1
|
Moreno-Estar S, Cidad P, Arevalo-Martinez M, Portillo AM, Sacristan-Moraleda M, Alonso E, Lopez-Lopez JR, Perez-Garcia MT. Vascular Smooth Muscle Cell Migration and P70S6K: Key Players in Intimal Hyperplasia Development. J Am Heart Assoc 2025; 14:e038358. [PMID: 40314369 DOI: 10.1161/jaha.124.038358] [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: 08/15/2024] [Accepted: 03/19/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Vascular smooth muscle cell (VSMC) recruitment and activation by vessel injury cause intimal hyperplasia (IH) and restenosis. Drug-eluting stents releasing mTOR (mechanistic target of rapamycin) blockers (sirolimus, everolimus [EV]) improve surgery outcomes but exhibit nonspecific effects and poor efficacy in diseased vessels. Drug combinations targeting the multifactorial processes leading to IH could enhance efficacy and reduce toxicity. Our previous work showed that Kv1.3 channel blockers such as 5-(4-phenoxybutoxy)psoralen (PAP-1) prevented IH. Since Kv1.3 signaling works through the MEK/ERK pathway, we hypothesize that PAP-1 and EV combination could improve antirestenotic therapies. METHODS AND RESULTS The effects of PAP-1, EV, and their combination on IH development were studied in vivo using a carotid ligation mouse model and ex vivo in organ culture of human vessels. Individually, both drugs inhibited vessel remodeling, but, surprisingly, their combination canceled these inhibitory effects. In primary human VSMCs cultures, the drug combination abolished the inhibition of cell migration but not cell proliferation, which was even potentiated. We uncovered a crosstalk between mTOR and MEK/ERK pathways in VSMCs, centered on P70S6K activation. P70S6K phosphorylation levels correlated with IH development, even reproducing the differences in EV response between diabetic and nondiabetic samples. CONCLUSIONS VSMC migration, rather than proliferation, mirrors PAP-1 and EV effects on IH development in vessels. Critically, we identify VSMC P70S6K phosphorylation as a surrogate marker for IH progression. The nonmonotonic responses of P70S6K activation to pathway blockers suggest the existence of a crosstalk element functioning as an exclusive NOR logic gate providing new insights for IH prevention strategies.
Collapse
MESH Headings
- Animals
- Ribosomal Protein S6 Kinases, 70-kDa/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Cell Movement/drug effects
- Hyperplasia
- Humans
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Everolimus/pharmacology
- Disease Models, Animal
- Neointima
- Mice, Inbred C57BL
- Cell Proliferation/drug effects
- Cells, Cultured
- Mice
- Male
- TOR Serine-Threonine Kinases/metabolism
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- Signal Transduction
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/enzymology
- Phosphorylation
- Vascular Remodeling/drug effects
- MTOR Inhibitors/pharmacology
Collapse
Affiliation(s)
- Sara Moreno-Estar
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| | - Pilar Cidad
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| | - Marycarmen Arevalo-Martinez
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| | - Ana M Portillo
- Departamento de Matemática Aplicada Universidad de Valladolid Instituto de Investigación en Matemáticas Valladolid Spain
| | - Marcos Sacristan-Moraleda
- Departamento de Matemática Aplicada Universidad de Valladolid Instituto de Investigación en Matemáticas Valladolid Spain
| | - Esperanza Alonso
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| | - Jose R Lopez-Lopez
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| | - M Teresa Perez-Garcia
- Departamento de Bioquímica y Biología Molecular y Fisiología Universidad de Valladolid Valladolid Spain
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), CSIC Valladolid Spain
| |
Collapse
|
2
|
Takiguchi K, Yokoi K, Murase D, Yokota M, Kawabata K, Takahashi Y, Minami S, Nakamura S, Yoshimori T, Watanabe R, Fujimoto M, Tanemura A. Significant Role of Autophagy in Melanosomal Degradation of Dermal Macrophages: Therapeutic Insight Regarding Hyperpigmentation with Uncertain Etiology. J Invest Dermatol 2025; 145:1229-1233.e6. [PMID: 39332604 DOI: 10.1016/j.jid.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 09/29/2024]
Affiliation(s)
| | - Kazunori Yokoi
- Department of Dermatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Daiki Murase
- Biological Science Research, Kao, Odawara, Japan.
| | | | | | | | - Satoshi Minami
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Shuhei Nakamura
- Department of Biochemistry, Nara Medical University, Kashihara, Japan
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Rei Watanabe
- Department of Dermatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
3
|
Hsu CY, Jasim SA, Bansal P, Kaur H, Ahmad I, Saud A, Deorari M, Al-Mashhadani ZI, Kumar A, Zwamel AH. Delving Into lncRNA-Mediated Regulation of Autophagy-Associated Signaling Pathways in the Context of Breast Cancer. Cell Biol Int 2025; 49:221-234. [PMID: 39873206 DOI: 10.1002/cbin.12277] [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: 05/11/2024] [Revised: 12/15/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
Breast cancer is a multifaceted and prevalent malignancy, impacting a considerable proportion of women globally. Numerous signaling pathways intricately regulate cellular functions such as growth, proliferation, and survival. Among the various regulators, lncRNAs have emerged as significant players despite their inability to encode proteins. An expanding body of literature underscores the pivotal roles lncRNAs play in cancer biology, particularly in the context of breast cancer. Autophagy, the cellular process dedicated to the degradation and recycling of cellular components, is now recognized as a crucial factor in cancer initiation and progression. The interplay between lncRNAs, various signaling pathways, and autophagy in the pathophysiology of breast cancer remains an active area of investigation. Researchers have identified specific lncRNAs that are dysregulated in breast cancer patients, influencing the modulation of key signaling pathways. Using experimental methodologies and bioinformatics approaches, multiple lncRNAs have been elucidated, providing deeper insights into their contributions to breast cancer pathogenesis and metastatic processes. In summary, the pathophysiological landscape of breast cancer is characterized by the complex interactions involving lncRNA-mediated autophagy. This understanding paves the way for identifying novel therapeutic targets, prognostic markers, and diagnostic markers, ultimately contributing to improved treatment outcomes in breast cancer management.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University, Tempe Campus, Phoenix, Arizona, USA
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, College of Health and Medical Technology, Al-Maarif University College, Anbar, Iraq
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Abdulnaser Saud
- Department of Pharmacy, Al-Hadi University College, Baghdad, Iraq
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named After the First President of Russia Boris Yeltsin, Ekaterinburg, Russia
- Department of Mechanical Engineering, Karpagam Academy of Higher Education, Coimbatore, India
| | - Ahmed Hussein Zwamel
- Department of Medical Laboratory Technology, College of Medical Technology, The Islamic University, Najaf, Iraq
| |
Collapse
|
4
|
Gulieva RE, Ahmadvand P, Freedman BS. A novel rapalog shows improved safety vs. efficacy in a human organoid model of polycystic kidney disease. Stem Cell Reports 2025; 20:102395. [PMID: 39855202 PMCID: PMC11864154 DOI: 10.1016/j.stemcr.2024.102395] [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: 03/11/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/27/2025] Open
Abstract
The mammalian target of rapamycin (mTOR) pathway is a therapeutic target in polycystic kidney disease (PKD), but mTOR inhibitors such as everolimus have failed to show efficacy at tolerated doses in clinical trials. Here, we introduce AV457, a novel rapalog developed to reduce side effects, and assess its dose-dependent safety and efficacy versus everolimus in PKD1-/- and PKD2-/- human kidney organoids, which form cysts in a PKD-specific way. Both AV457 and everolimus reduce cyst growth over time. At intermediate doses, AV457 exhibits an improved safety profile relative to everolimus, with comparable efficacy. Target engagement assays confirm mTOR pathway inhibition and greater selectivity of AV457 for mTOR complex 1 versus complex 2, compared to everolimus. AV457 thus provides a more favorable balance of safety and efficacy for PKD compared to everolimus and merits further consideration as an investigational therapeutic.
Collapse
Affiliation(s)
- Ramila E Gulieva
- Department of Medicine, Division of Nephrology, Institute for Stem Cell & Regenerative Medicine, and Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA
| | | | - Benjamin S Freedman
- Department of Medicine, Division of Nephrology, Institute for Stem Cell & Regenerative Medicine, and Kidney Research Institute, University of Washington School of Medicine, Seattle, WA 98109, USA; Plurexa LLC, Seattle, WA 98109, USA.
| |
Collapse
|
5
|
Kumar D, Kanchan R, Chaturvedi NK. Targeting protein synthesis pathways in MYC-amplified medulloblastoma. Discov Oncol 2025; 16:23. [PMID: 39779613 PMCID: PMC11711608 DOI: 10.1007/s12672-025-01761-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
MYC is one of the most deregulated oncogenic transcription factors in human cancers. MYC amplification/or overexpression is most common in Group 3 medulloblastoma and is positively associated with poor prognosis. MYC is known to regulate the transcription of major components of protein synthesis (translation) machinery, leading to promoted rates of protein synthesis and tumorigenesis. MTOR signaling-driven deregulated protein synthesis is widespread in various cancers, including medulloblastoma, which can promote the stabilization of MYC. Indeed, our previous studies demonstrate that the key components of protein synthesis machinery, including mTOR signaling and MYC targets, are overexpressed and activated in MYC-amplified medulloblastoma, confirming MYC-dependent addiction of enhanced protein synthesis in medulloblastoma. Further, targeting this enhanced protein synthesis pathway with combined inhibition of MYC transcription and mTOR translation by small-molecule inhibitors, demonstrates preclinical synergistic anti-tumor potential against MYC-driven medulloblastoma in vitro and in vivo. Thus, inhibiting enhanced protein synthesis by targeting the MYC indirectly and mTOR pathways together may present a highly appropriate strategy for treating MYC-driven medulloblastoma and other MYC-addicted cancers. Evidence strongly proposes that MYC/mTOR-driven tumorigenic signaling can predominantly control the translational machinery to elicit cooperative effects on increased cell proliferation, cell cycle progression, and genome dysregulation as a mechanism of cancer initiation. Several small molecule inhibitors of targeting MYC indirectly and mTOR signaling have been developed and used clinically with immunosuppressants and chemotherapy in multiple cancers. Only a few of them have been investigated as treatments for medulloblastoma and other pediatric tumors. This review explores concurrent targeting of MYC and mTOR signaling against MYC-driven medulloblastoma. Based on existing evidence, targeting of MYC and mTOR pathways together produces functional synergy that could be the basis for effective therapies against medulloblastoma.
Collapse
Affiliation(s)
- Devendra Kumar
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 986395, USA
| | - Ranjana Kanchan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nagendra K Chaturvedi
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, 986395, USA.
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
6
|
Jian W, Zhang L. POLE2 silencing inhibits the progression of colorectal carcinoma cells via wnt signaling axis. Cancer Biol Ther 2024; 25:2392339. [PMID: 39155507 PMCID: PMC11340749 DOI: 10.1080/15384047.2024.2392339] [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: 12/27/2023] [Revised: 08/06/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant carcinoma worldwide. DNA polymerase epsilon 2, accessory subunit (POLE2) participates in DNA replication, repair, and cell cycle control, but its association with CRC development remains unclear. In the present study, the differentially expressed genes (DEGs) in CRC were screened from bioinformatics analysis based on GEO database. RT-qPCR was used to assess mRNA expression. CCK-8 and colony formation assays were applied for the evaluation of cell proliferation. Wound healing and transwell assays were used to detect cell migration and invasion. Protein levels were determined by Western blotting assay. We found that POLE2 was highly expressed in CRC tissues and cell lines. Inhibition of POLE2 suppressed the proliferation, migration and invasion of CRC cells. Mechanistically, Wnt/β-catenin signaling pathway was inactivated by inhibition of POLE2. Activation of Wnt/β-catenin pathway can reverse the function of POLE2 knockdown on CRC cells. In vivo studies demonstrated that POLE2 silencing could notably inhibit the growth of tumors, which was consistent with the results in vitro. In conclusion, we found POLE2 as a novel oncogene in CRC, providing a potential therapeutic or diagnostic target in CRC.
Collapse
Affiliation(s)
- Weihua Jian
- Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
- Department of General Surgery, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lei Zhang
- Department of Second General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, China
| |
Collapse
|
7
|
Du Y, Liu T, Ding T, Zeng X, Chen Q, Zhao H. Adhesive lipophilic gels delivering rapamycin prevent oral leukoplakia from malignant transformation. Mater Today Bio 2024; 29:101305. [PMID: 39525395 PMCID: PMC11546665 DOI: 10.1016/j.mtbio.2024.101305] [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: 09/03/2024] [Revised: 10/14/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024] Open
Abstract
Oral leukoplakia (OLK) is the most emblematic oral potentially malignant disorder that may precede the diagnosis of oral squamous cell carcinoma (OSCC) and has an overall malignant transformation rate of 9.8 %. Early intervention is crucial to reduce the malignant transformation rate from OLK to OSCC but the lack of effective local pharmaceutical preparations poses a challenge to clinical management. Rapamycin is speculated to prevent OLK from carcinogenesis and its inherent lipophilicity facilitates its penetration into stratum corneum. Nevertheless, hydrophilic hydrogels frequently encounter challenges when attempting to deliver lipophilic drugs. Furthermore, the oral cavity presents a complex environment defined by oral motor functions, saliva secretion cycles, dynamic fluctuations, and protective barriers comprising mucus and lipid layers. Consequently, addressing issues of muco-penetration and muco-adhesion is imperative for developing an effective drug delivery system aiming at delivering rapamycin to target oral potentially malignant disorders. Here, a dual-function hydrogel drug delivery system integrating adhesion and lipophilicity was successfully developed based on polyvinyl alcohol (PVA) and dioleoyl phosphatidylglycerol (DOPG) via dynamic boronic ester bonds. Rheological experiments based on orthogonal design revealed that PVA-DOPG hydrogels exhibited ideal adhesive strength (around 6 kPa) and could adhere to various surfaces in both dry and wet conditions. PVA-DOPG hydrogels also significantly promoted lipophilic molecules' penetration into stratum corneum (integrated fluorescence density of 6.95 ± 0.52 × 106 and mean fluorescence depth of 0.96 ± 0.07 mm) of ex-vivo porcine buccal mucosa (p < 0.001). Furthermore, PVA-DOPG hydrogels incorporating rapamycin inhibited malignant transformation of OLK mouse model induced by 4-Nitroquinoline N-oxide (4-NQO), distinct improvements in survival (the neoplasm incidence density at the 40th day is 0.0091) (p < 0.05), decrease in neoplasm incidence density of 36.36 % and inhibition rate in neoplasm volume of 75.04 ± 33.67 % have been demonstrated, suggesting the hydrogels were valuable candidates for potential applications in the management of OLK.
Collapse
Affiliation(s)
- Yuqi Du
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, PR China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Med-X Center for Materials, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Tiannan Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Med-X Center for Materials, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Tingting Ding
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, PR China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Med-X Center for Materials, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, PR China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Med-X Center for Materials, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, PR China
| |
Collapse
|
8
|
Li X, Yang C, Zhang X, Wang F, Sun L, Zhang W, Xu X. R-loop formation contributes to mTORC1 activation-dependent DNA replication stress induced by p53 deficiency. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1875-1885. [PMID: 39592262 PMCID: PMC11693875 DOI: 10.3724/abbs.2024188] [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: 10/20/2023] [Accepted: 07/30/2024] [Indexed: 11/28/2024] Open
Abstract
DNA replication stress is a significant contributor to spontaneous DNA damage and genome instability. While the impact of p53 deficiency on increasing DNA replication stress is known, the specific molecular mechanism underlying this phenomenon remains poorly understood. This study explores how p53 deficiency induces DNA replication stress by activating mTORC1 through R-loop formation, which is facilitated by the upregulation of RNR. Research has shown that p53 deficiency results in increased γH2AX expression and a higher mutation rate in the HPRT gene. Interestingly, these effects can be alleviated by rapamycin, an mTORC1 inhibitor. Additionally, rapamycin reduces the abundance of R-loop structures in p53KO cells, which is linked to mTORC1's regulation of ribonucleotide reductase (RNR) level. These findings suggest that p53 deficiency-induced DNA replication stress relies on mTORC1 activation, with the upregulation of RNR expression and R-loop formation. Overall, this study underscores the importance of R-loops in mTORC1 activation-dependent DNA replication stress triggered by p53 deficiency.
Collapse
Affiliation(s)
- Xiaolei Li
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- China-Japan Friendship Jiangxi HospitalNational Regional Center for Respiratory MedicineNanchang330200China
| | - Cheng Yang
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Jiangxi Medical CollegeFirst Clinical Medical CollegeNanchang UniversityNanchang330006China
| | - Xiaohui Zhang
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- Department of Respiratory and Critical Care MedicineRenmin Hospital of ShangraoShangrao334000China
| | - Feiyang Wang
- Jiangxi Medical CollegeFirst Clinical Medical CollegeNanchang UniversityNanchang330006China
| | - Longhua Sun
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- China-Japan Friendship Jiangxi HospitalNational Regional Center for Respiratory MedicineNanchang330200China
| | - Wei Zhang
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- China-Japan Friendship Jiangxi HospitalNational Regional Center for Respiratory MedicineNanchang330200China
| | - Xinping Xu
- Jiangxi Provincial Key Laboratory of Respiratory DiseasesJiangxi Institute of Respiratory DiseaseDepartment of Respiratory and Critical Care Medicinethe First Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchang330006China
- China-Japan Friendship Jiangxi HospitalNational Regional Center for Respiratory MedicineNanchang330200China
| |
Collapse
|
9
|
Matteini F, Montserrat‐Vazquez S, Florian MC. Rejuvenating aged stem cells: therapeutic strategies to extend health and lifespan. FEBS Lett 2024; 598:2776-2787. [PMID: 38604982 PMCID: PMC11586596 DOI: 10.1002/1873-3468.14865] [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: 10/23/2023] [Revised: 02/03/2024] [Accepted: 03/07/2024] [Indexed: 04/13/2024]
Abstract
Aging is associated with a global decline in stem cell function. To date, several strategies have been proposed to rejuvenate aged stem cells: most of these result in functional improvement of the tissue where the stem cells reside, but the impact on the lifespan of the whole organism has been less clearly established. Here, we review some of the most recent work dealing with interventions that improve the regenerative capacity of aged somatic stem cells in mammals and that might have important translational possibilities. Overall, we underscore that somatic stem cell rejuvenation represents a strategy to improve tissue homeostasis upon aging and present some recent approaches with the potential to affect health span and lifespan of the whole organism.
Collapse
Affiliation(s)
- Francesca Matteini
- Stem Cell Aging Group, Regenerative Medicine ProgramThe Bellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P‐CMR[C])BarcelonaSpain
| | - Sara Montserrat‐Vazquez
- Stem Cell Aging Group, Regenerative Medicine ProgramThe Bellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P‐CMR[C])BarcelonaSpain
| | - M. Carolina Florian
- Stem Cell Aging Group, Regenerative Medicine ProgramThe Bellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P‐CMR[C])BarcelonaSpain
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN)MadridSpain
- The Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
| |
Collapse
|
10
|
Ghosh R, Bishayi B. Neutralization of TLR2 in combination with either TNF-α or IL-1β antibody reduces the severity of septic arthritis through STAT3/mTOR signalling in lymphocytes. Cell Immunol 2024; 405-406:104878. [PMID: 39312873 DOI: 10.1016/j.cellimm.2024.104878] [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: 07/06/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024]
Abstract
Staphylococcus aureus induced Septic arthritis is considered a medical concern. S.aureus binds TLR2 to induce an array of inflammatory responses. Generation of pro-inflammatory cytokines induces T cell responses and control Th17/Treg cell balance. Regulation of T cell-mediated immunity in response to inflammation is significantly influenced by mTOR. Presence of elevated TNF-α, IL-1β decreases Treg cell activity through STAT3/mTOR, promoting proliferation of T cells towards Th17 cells. Therefore, we postulated, neutralizing TLR2 with either TNF-α or IL-1β in combination could be useful in modifying Th17/Treg cell ratio in order to treat septic arthritis by suppressing expression of mTOR/STAT3. To date, no studies have reported effects of neutralization of TLR2 along with either TNF-α or IL-1β on amelioration of arthritis correlating with mTOR/STAT3 expression. Contribution of T lymphocytes collected from blood, spleen, synovial tissues, their derived cytokines IFN-γ, IL-6, IL-17, TGF-β, IL-10 were noted. Expression of TLR2, TNFR1, TNFR2, NF-κB along with mTOR/STAT3 also recorded. Neutralization of TLR2 along with TNF-α and IL-1β were able to shift Th17 cells into immunosuppressive Treg cells. Furthermore,elevated expression of IL-10, TNFR2 and demoted expression of mTOR/ STAT3 along with NF-κB in lymphocytes confirms its role in resolution of arthritis. It was also effective in reducing oxidative stress via increasing expression of the antioxidant enzymes. As a result, it can be inferred that Treg-derived IL-10, which may mitigate inflammatory effects of septic arthritis by influencing the mTOR/STAT3 interaction in lymphocytes, may be selected as a different therapeutic strategy for reducing the impact of septic arthritis.
Collapse
Affiliation(s)
- Rituparna Ghosh
- Department of Physiology, Immunology laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India.
| | - Biswadev Bishayi
- Department of Physiology, Immunology laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India.
| |
Collapse
|
11
|
Barmaki H, Nourazarian A, Shademan B, Khaki-Khatibi F. The autophagy paradox: A new hypothesis in neurodegenerative disorders. Neurochem Int 2024; 179:105827. [PMID: 39111406 DOI: 10.1016/j.neuint.2024.105827] [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: 05/22/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
A recent study showed that while autophagy is usually tied to protein and organelle turnover, it can also play dual roles in neurodegenerative diseases. Traditionally, autophagy was seen as protective since it removes damaged proteins and organelles. but new data suggests autophagy can sometimes promote neuron death. and This review tackles autophagy's seemingly contradictory effects in neurodegeneration, or the "autophagy paradox. " It offers a framework for understanding autophagy in neurodegenerative research and the cellular processes involved. In short, our data uncovers a harmful autophagy role in certain situations, conflicting the view that it's always beneficial. We describe the distinct, disease-specific autophagy pathways functioning in various neurodegenerative diseases. Part two concerns potential therapeutic implications of manipulating autophagy and current strategies targeting the autophagic system, suggesting interesting areas for future research into tailored modulators. This could eventually enable activating or controlling specific autophagy pathways and aid in developing more effective treatments. Researchers believe more molecular-level research is needed so patient-tailored autophagy-modulating therapeutics can be developed given this dilemma. Moreover, research must translate faster into effective neurodegenerative disease treatment options. This article aims to provide a wholly new perspective on autophagy's classically described role in these severe diseases, challenging current dogma and opening new therapeutic avenue options.
Collapse
Affiliation(s)
- Haleh Barmaki
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran; Student Research Committee, Khoy University of Medical Sciences, Khoy, Iran.
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Khaki-Khatibi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
12
|
Ma L, Yu J, Fu Y, He X, Ge S, Jia R, Zhuang A, Yang Z, Fan X. The dual role of cellular senescence in human tumor progression and therapy. MedComm (Beijing) 2024; 5:e695. [PMID: 39161800 PMCID: PMC11331035 DOI: 10.1002/mco2.695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/21/2024] Open
Abstract
Cellular senescence, one of the hallmarks of cancer, is characterized by cell cycle arrest and the loss of most normal cellular functions while acquiring a hypersecretory, proinflammatory phenotype. The function of senescent cells in cancer cells varies depending on the cellular conditions. Before the occurrence of cancer, senescent cells act as a barrier to prevent its development. But once cancer has occurred, senescent cells play a procancer role. However, few of the current studies have adequately explained the diversity of cellular senescence across cancers. Herein, we concluded the latest intrinsic mechanisms of cellular senescence in detail and emphasized the senescence-associated secretory phenotype as a key contributor to heterogeneity of senescent cells in tumor. We also discussed five kinds of inducers of cellular senescence and the advancement of senolytics in cancer, which are drugs that tend to clear senescent cells. Finally, we summarized the various effects of senescent cells in different cancers and manifested that their functions may be diametrically opposed under different circumstances. In short, this paper contributes to the understanding of the diversity of cellular senescence in cancers and provides novel insight for tumor therapy.
Collapse
Affiliation(s)
- Liang Ma
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Jie Yu
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Yidian Fu
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Xiaoyu He
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Shengfang Ge
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Renbing Jia
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Ai Zhuang
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Zhi Yang
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Xianqun Fan
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| |
Collapse
|
13
|
Zhang WY, Wei QQ, Zhang T, Wang CS, Chen J, Wang JH, Xie X, Jiang P. Microglial AKAP8L: a key mediator in diabetes-associated cognitive impairment via autophagy inhibition and neuroinflammation triggering. J Neuroinflammation 2024; 21:177. [PMID: 39033121 PMCID: PMC11264944 DOI: 10.1186/s12974-024-03170-z] [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: 04/24/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Diabetes-associated cognitive impairment (DACI) poses a significant challenge to the self-management of diabetes, markedly elevating the risk of adverse complications. A burgeoning body of evidence implicates microglia as a central player in the pathogenesis of DACI. METHODS We utilized proteomics to identify potential biomarkers in high glucose (HG)-treated microglia, followed by gene knockdown techniques for mechanistic validation in vitro and in vivo. RESULTS Our proteomic analysis identified a significant upregulation of AKAP8L in HG-treated microglia, with concurrent dysregulation of autophagy and inflammation markers, making AKAP8L a novel biomarker of interest. Notably, the accumulation of AKAP8L was specific to HG-treated microglia, with no observed changes in co-cultured astrocytes or neurons, a pattern that was mirrored in streptozotocin (STZ)-induced diabetic mice. Further studies through co-immunoprecipitation and proximity ligation assay indicated that the elevated AKAP8L in HG-treated microglial cells interacts with the mTORC1. In the STZ mouse model, we demonstrated that both AKAP8L knockdown and rapamycin treatment significantly enhanced cognitive function, as evidenced by improved performance in the Morris water maze, and reduced microglial activation. Moreover, these interventions effectively suppressed mTORC1 signaling, normalized autophagic flux, mitigated neuroinflammation, and decreased pyroptosis. CONCLUSIONS Our findings highlight the critical role of AKAP8L in the development of DACI. By interacting with mTORC1, AKAP8L appears to obstruct autophagic processes and initiate a cascade of neuroinflammatory responses. The identification of AKAP8L as a key mediator in DACI opens up new avenues for potential therapeutic interventions.
Collapse
Affiliation(s)
- Wen-Yuan Zhang
- Department of Pharmacy, Zhongshan City People's Hospital, Zhongshan, 528403, China
- School of Pharmaceutical Sciences, Zunyi Medical University, Zunyi, 510006, China
| | - Qian-Qian Wei
- Department of Pharmacy, Zhongshan City People's Hospital, Zhongshan, 528403, China
- School of Pharmaceutical Sciences, Zunyi Medical University, Zunyi, 510006, China
| | - Tao Zhang
- Translational Pharmaceutical Laboratory, Jining First People ' s Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Chang-Shui Wang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Jing Chen
- Neurobiology Key Laboratory, Jining Medical University, Jining, 272067, China
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Jian-Hua Wang
- Translational Pharmaceutical Laboratory, Jining First People ' s Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Xin Xie
- Translational Pharmaceutical Laboratory, Jining First People ' s Hospital, Shandong First Medical University, Jining, 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People ' s Hospital, Shandong First Medical University, Jining, 272000, China.
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, 272000, China.
| |
Collapse
|
14
|
Taeb S, Rostamzadeh D, Amini SM, Rahmati M, Eftekhari M, Safari A, Najafi M. MicroRNAs targeted mTOR as therapeutic agents to improve radiotherapy outcome. Cancer Cell Int 2024; 24:233. [PMID: 38965615 PMCID: PMC11229485 DOI: 10.1186/s12935-024-03420-3] [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: 11/03/2023] [Accepted: 06/22/2024] [Indexed: 07/06/2024] Open
Abstract
MicroRNAs (miRNAs) are small RNA molecules that regulate genes and are involved in various biological processes, including cancer development. Researchers have been exploring the potential of miRNAs as therapeutic agents in cancer treatment. Specifically, targeting the mammalian target of the rapamycin (mTOR) pathway with miRNAs has shown promise in improving the effectiveness of radiotherapy (RT), a common cancer treatment. This review provides an overview of the current understanding of miRNAs targeting mTOR as therapeutic agents to enhance RT outcomes in cancer patients. It emphasizes the importance of understanding the specific miRNAs that target mTOR and their impact on radiosensitivity for personalized cancer treatment approaches. The review also discusses the role of mTOR in cell homeostasis, cell proliferation, and immune response, as well as its association with oncogenesis. It highlights the different ways in which miRNAs can potentially affect the mTOR pathway and their implications in immune-related diseases. Preclinical findings suggest that combining mTOR modulators with RT can inhibit tumor growth through anti-angiogenic and anti-vascular effects, but further research and clinical trials are needed to validate the efficacy and safety of using miRNAs targeting mTOR as therapeutic agents in combination with RT. Overall, this review provides a comprehensive understanding of the potential of miRNAs targeting mTOR to enhance RT efficacy in cancer treatment and emphasizes the need for further research to translate these findings into improved clinical outcomes.
Collapse
Affiliation(s)
- Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Davoud Rostamzadeh
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Seyed Mohammad Amini
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Rahmati
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Eftekhari
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Arash Safari
- Department of Radiology, Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, 71439-14693, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Medical Biology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
15
|
Spanjaard P, Petit JM, Schmitt A, Vergès B, Bouillet B. Screening and management of metabolic complications of mTOR inhibitors in real-life settings. ANNALES D'ENDOCRINOLOGIE 2024; 85:263-268. [PMID: 38043912 DOI: 10.1016/j.ando.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/10/2023] [Accepted: 11/20/2023] [Indexed: 12/05/2023]
Abstract
INTRODUCTION Inhibitors of mTOR (mTORi) are frequently used as anticancer treatment. They were responsible for metabolic side-effects in phase 3 studies, which provided only an incomplete picture of these metabolic complications. The aim of our study was therefore to evaluate, in a real-life setting, outcomes for patients with dyslipidemia or diabetes under mTORi, and the incidence and management of metabolic abnormalities occurring under mTORi in the absence of known metabolic history. METHODS This single-center retrospective study included all 177 patients receiving everolimus in the Cancer Center of Dijon, France, between May 2015 and November 2018. RESULTS Diabetes was diagnosed in 15 patients (9%), with an estimated mean time to onset of 160±173 days. Antidiabetic treatment was introduced in 41% of these patients. After mTORi discontinuation, diabetes persisted in 60% of patients in whom it had been diagnosed. Dyslipidemia was diagnosed in 22 patients (14%): 55% with hypercholesterolemia and 45% with hypertriglyceridemia. 18% were placed on lipid-lowering therapy. While all patients were screened for hyperglycemia and monitored for known diabetes, only 42% of patients without dyslipidemia were screened for lipids, and only 8% of patients with known dyslipidemia were monitored for lipids. CONCLUSION Our study is one of the few to look at metabolic complications secondary to mTORi in a real-life situation. The incidence of diabetes was high, but the use of antidiabetic treatment was variable. Normalization of glucose homeostasis after mTORi discontinuation is possible, particularly in patients who have not been placed on antidiabetic therapy. Screening for dyslipidemia was clearly inadequate in our study, making the data on this point more difficult to interpret. It appears that adherence to guidelines needs to be improved to optimize the management of patients treated with mTORi.
Collapse
Affiliation(s)
- Pamela Spanjaard
- Department of Endocrinology, Diabetes and Metabolic Disorders, Dijon University Hospital, Dijon, France
| | - Jean Michel Petit
- Department of Endocrinology, Diabetes and Metabolic Disorders, Dijon University Hospital, Dijon, France; Inserm Unit, LNC-UMR 1231, University of Burgundy, Dijon, France
| | - Antonin Schmitt
- Inserm Unit, LNC-UMR 1231, University of Burgundy, Dijon, France; Department of Pharmacy, Centre Georges-François Leclerc, Dijon, France
| | - Bruno Vergès
- Department of Endocrinology, Diabetes and Metabolic Disorders, Dijon University Hospital, Dijon, France; Inserm Unit, LNC-UMR 1231, University of Burgundy, Dijon, France
| | - Benjamin Bouillet
- Department of Endocrinology, Diabetes and Metabolic Disorders, Dijon University Hospital, Dijon, France; Inserm Unit, LNC-UMR 1231, University of Burgundy, Dijon, France.
| |
Collapse
|
16
|
Chen J, Gao Y, Liu N, Hai D, Wei W, Liu Y, Lan X, Jin X, Yu J, Ma L. Mechanism of NLRP3 Inflammasome in Epilepsy and Related Therapeutic Agents. Neuroscience 2024; 546:157-177. [PMID: 38574797 DOI: 10.1016/j.neuroscience.2024.03.029] [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: 12/31/2023] [Revised: 03/05/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024]
Abstract
Epilepsy is one of the most widespread and complex diseases in the central nervous system (CNS), affecting approximately 65 million people globally, an important factor resulting in neurological disability-adjusted life year (DALY) and progressive cognitive dysfunction. Medication is the most essential treatment. The currently used drugs have shown drug resistance in some patients and only control symptoms; the development of novel and more efficacious pharmacotherapy is imminent. Increasing evidence suggests neuroinflammation is involved in the occurrence and development of epilepsy, and high expression of NLRP3 inflammasome has been observed in the temporal lobe epilepsy (TLE) brain tissue of patients and animal models. The inflammasome is a crucial cause of neuroinflammation by activating IL-1β and IL-18. Many preclinical studies have confirmed that regulating NLRP3 inflammasome pathway can prevent the development of epilepsy, reduce the severity of epilepsy, and play a neuroprotective role. Therefore, regulating NLRP3 inflammasome could be a potential target for epilepsy treatment. In summary, this review describes the priming and activation of inflammasome and its biological function in the progression of epilepsy. In addition, we reviewes the current pharmacological researches for epilepsy based on the regulation of NLRP3 inflammasome, aiming to provide a basis and reference for developing novel antiepileptic drugs.
Collapse
Affiliation(s)
- Juan Chen
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Yuan Gao
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Ning Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Dongmei Hai
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Wei Wei
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Yue Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Xiaobing Lan
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Xueqin Jin
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China.
| | - Jianqiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China.
| | - Lin Ma
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
17
|
Perurena N, Situ L, Cichowski K. Combinatorial strategies to target RAS-driven cancers. Nat Rev Cancer 2024; 24:316-337. [PMID: 38627557 DOI: 10.1038/s41568-024-00679-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2024] [Indexed: 05/01/2024]
Abstract
Although RAS was formerly considered undruggable, various agents that inhibit RAS or specific RAS oncoproteins have now been developed. Indeed, the importance of directly targeting RAS has recently been illustrated by the clinical success of mutant-selective KRAS inhibitors. Nevertheless, responses to these agents are typically incomplete and restricted to a subset of patients, highlighting the need to develop more effective treatments, which will likely require a combinatorial approach. Vertical strategies that target multiple nodes within the RAS pathway to achieve deeper suppression are being investigated and have precedence in other contexts. However, alternative strategies that co-target RAS and other therapeutic vulnerabilities have been identified, which may mitigate the requirement for profound pathway suppression. Regardless, the efficacy of any given approach will likely be dictated by genetic, epigenetic and tumour-specific variables. Here we discuss various combinatorial strategies to treat KRAS-driven cancers, highlighting mechanistic concepts that may extend to tumours harbouring other RAS mutations. Although many promising combinations have been identified, clinical responses will ultimately depend on whether a therapeutic window can be achieved and our ability to prospectively select responsive patients. Therefore, we must continue to develop and understand biologically diverse strategies to maximize our likelihood of success.
Collapse
Affiliation(s)
- Naiara Perurena
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Lisa Situ
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Karen Cichowski
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Ludwig Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Allard C, Miralpeix C, López-Gambero AJ, Cota D. mTORC1 in energy expenditure: consequences for obesity. Nat Rev Endocrinol 2024; 20:239-251. [PMID: 38225400 DOI: 10.1038/s41574-023-00934-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2023] [Indexed: 01/17/2024]
Abstract
In eukaryotic cells, the mammalian target of rapamycin complex 1 (sometimes referred to as the mechanistic target of rapamycin complex 1; mTORC1) orchestrates cellular metabolism in response to environmental energy availability. As a result, at the organismal level, mTORC1 signalling regulates the intake, storage and use of energy by acting as a hub for the actions of nutrients and hormones, such as leptin and insulin, in different cell types. It is therefore unsurprising that deregulated mTORC1 signalling is associated with obesity. Strategies that increase energy expenditure offer therapeutic promise for the treatment of obesity. Here we review current evidence illustrating the critical role of mTORC1 signalling in the regulation of energy expenditure and adaptive thermogenesis through its various effects in neuronal circuits, adipose tissue and skeletal muscle. Understanding how mTORC1 signalling in one organ and cell type affects responses in other organs and cell types could be key to developing better, safer treatments targeting this pathway in obesity.
Collapse
Affiliation(s)
- Camille Allard
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | | | | | - Daniela Cota
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France.
| |
Collapse
|
19
|
Appelbaum J, Price AE, Oda K, Zhang J, Leung WH, Tampella G, Xia D, So PP, Hilton SK, Evandy C, Sarkar S, Martin U, Krostag AR, Leonardi M, Zak DE, Logan R, Lewis P, Franke-Welch S, Ngwenyama N, Fitzgerald M, Tulberg N, Rawlings-Rhea S, Gardner RA, Jones K, Sanabria A, Crago W, Timmer J, Hollands A, Eckelman B, Bilic S, Woodworth J, Lamble A, Gregory PD, Jarjour J, Pogson M, Gustafson JA, Astrakhan A, Jensen MC. Drug-regulated CD33-targeted CAR T cells control AML using clinically optimized rapamycin dosing. J Clin Invest 2024; 134:e162593. [PMID: 38502193 PMCID: PMC11060733 DOI: 10.1172/jci162593] [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: 06/13/2022] [Accepted: 03/08/2024] [Indexed: 03/21/2024] Open
Abstract
Chimeric antigen receptor (CAR) designs that incorporate pharmacologic control are desirable; however, designs suitable for clinical translation are needed. We designed a fully human, rapamycin-regulated drug product for targeting CD33+ tumors called dimerizaing agent-regulated immunoreceptor complex (DARIC33). T cell products demonstrated target-specific and rapamycin-dependent cytokine release, transcriptional responses, cytotoxicity, and in vivo antileukemic activity in the presence of as little as 1 nM rapamycin. Rapamycin withdrawal paused DARIC33-stimulated T cell effector functions, which were restored following reexposure to rapamycin, demonstrating reversible effector function control. While rapamycin-regulated DARIC33 T cells were highly sensitive to target antigen, CD34+ stem cell colony-forming capacity was not impacted. We benchmarked DARIC33 potency relative to CD19 CAR T cells to estimate a T cell dose for clinical testing. In addition, we integrated in vitro and preclinical in vivo drug concentration thresholds for off-on state transitions, as well as murine and human rapamycin pharmacokinetics, to estimate a clinically applicable rapamycin dosing schedule. A phase I DARIC33 trial has been initiated (PLAT-08, NCT05105152), with initial evidence of rapamycin-regulated T cell activation and antitumor impact. Our findings provide evidence that the DARIC platform exhibits sensitive regulation and potency needed for clinical application to other important immunotherapy targets.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Immunotherapy, Adoptive
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Receptors, Chimeric Antigen/immunology
- Sialic Acid Binding Ig-like Lectin 3/immunology
- Sialic Acid Binding Ig-like Lectin 3/metabolism
- Sirolimus/pharmacology
- Sirolimus/administration & dosage
- T-Lymphocytes/immunology
- T-Lymphocytes/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jacob Appelbaum
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
- Division of Hematology/Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
- Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Seattle Children’s Hospital, Seattle, Washington, USA
| | | | - Kaori Oda
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Joy Zhang
- 2seventy bio, Cambridge, Massachusetts, USA
| | | | - Giacomo Tampella
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Dong Xia
- 2seventy bio, Cambridge, Massachusetts, USA
| | | | | | - Claudya Evandy
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Semanti Sarkar
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | | | - Marissa Leonardi
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Rachael Logan
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | | | | | - Michael Fitzgerald
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Niklas Tulberg
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Stephanie Rawlings-Rhea
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Rebecca A. Gardner
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Kyle Jones
- Inhibrx, Torrey Pines Science Park, La Jolla, California, USA
| | | | - William Crago
- Inhibrx, Torrey Pines Science Park, La Jolla, California, USA
| | - John Timmer
- Inhibrx, Torrey Pines Science Park, La Jolla, California, USA
| | - Andrew Hollands
- Inhibrx, Torrey Pines Science Park, La Jolla, California, USA
| | | | | | | | - Adam Lamble
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
- Seattle Children’s Hospital, Seattle, Washington, USA
| | | | | | | | - Joshua A. Gustafson
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Michael C. Jensen
- Seattle Children’s Therapeutics, Seattle Children’s Research Institute, Seattle, Washington, USA
| |
Collapse
|
20
|
Cullen ER, Safari M, Mittelstadt I, Weston MC. Hyperactivity of mTORC1- and mTORC2-dependent signaling mediates epilepsy downstream of somatic PTEN loss. eLife 2024; 12:RP91323. [PMID: 38446016 PMCID: PMC10942640 DOI: 10.7554/elife.91323] [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] [Indexed: 03/07/2024] Open
Abstract
Gene variants that hyperactivate PI3K-mTOR signaling in the brain lead to epilepsy and cortical malformations in humans. Some gene variants associated with these pathologies only hyperactivate mTORC1, but others, such as PTEN, PIK3CA, and AKT, hyperactivate both mTORC1- and mTORC2-dependent signaling. Previous work established a key role for mTORC1 hyperactivity in mTORopathies, however, whether mTORC2 hyperactivity contributes is not clear. To test this, we inactivated mTORC1 and/or mTORC2 downstream of early Pten deletion in a new mouse model of somatic Pten loss-of-function (LOF) in the cortex and hippocampus. Spontaneous seizures and epileptiform activity persisted despite mTORC1 or mTORC2 inactivation alone, but inactivating both mTORC1 and mTORC2 simultaneously normalized brain activity. These results suggest that hyperactivity of both mTORC1 and mTORC2 can cause epilepsy, and that targeted therapies should aim to reduce activity of both complexes.
Collapse
Affiliation(s)
- Erin R Cullen
- Department of Neurological Sciences, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Mona Safari
- Fralin Biomedical Research Institute at VTC, Center for Neurobiology ResearchRoanokeUnited States
- Translational Biology, Medicine, and Health Graduate ProgramRoanokeUnited States
| | - Isabelle Mittelstadt
- Department of Neurological Sciences, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Matthew C Weston
- Department of Neurological Sciences, Larner College of Medicine, University of VermontBurlingtonUnited States
- Fralin Biomedical Research Institute at VTC, Center for Neurobiology ResearchRoanokeUnited States
- School of Neuroscience, Virginia Polytechnic and State UniversityBlacksburgUnited States
| |
Collapse
|
21
|
Petr MA, Matiyevskaya F, Osborne B, Berglind M, Reves S, Zhang B, Ezra MB, Carmona-Marin LM, Syadzha MF, Mediavilla MC, Keijzers G, Bakula D, Mkrtchyan GV, Scheibye-Knudsen M. Pharmacological interventions in human aging. Ageing Res Rev 2024; 95:102213. [PMID: 38309591 DOI: 10.1016/j.arr.2024.102213] [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: 08/09/2023] [Revised: 01/15/2024] [Accepted: 01/30/2024] [Indexed: 02/05/2024]
Abstract
Pharmacological interventions are emerging as potential avenues of alleviating age-related disease. However, the knowledge of ongoing clinical trials as they relate to aging and pharmacological interventions is dispersed across a variety of mediums. In this review we summarize 136 age-related clinical trials that have been completed or are ongoing. Furthermore, we establish a database that describe the trials (AgingDB, www.agingdb.com) keeping track of the previous and ongoing clinical trials, alongside their outcomes. The aim of this review and database is to give people the ability to easily query for their trial of interest and stay up to date on the latest results. In sum, herein we give an overview of the current pharmacological strategies that have been applied to target human aging.
Collapse
Affiliation(s)
- Michael Angelo Petr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Frida Matiyevskaya
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Brenna Osborne
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Magnus Berglind
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Simon Reves
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Bin Zhang
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Michael Ben Ezra
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Lina Maria Carmona-Marin
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Muhammad Farraz Syadzha
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Marta Cortés Mediavilla
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Guido Keijzers
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Garik V Mkrtchyan
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
22
|
Ankrah PK, Mensah ED, Dabie K, Mensah C, Akangbe B, Essuman J. Harnessing Genetics to Extend Lifespan and Healthspan: Current Progress and Future Directions. Cureus 2024; 16:e55495. [PMID: 38571872 PMCID: PMC10990068 DOI: 10.7759/cureus.55495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/05/2024] Open
Abstract
Aging is inevitable, but the lifespan (duration of life) and healthspan (healthy aging) vary greatly among individuals and across species. Unlocking the secrets behind these differences has captivated scientific curiosity for ages. This review presents relevant recent advances in genetics and cell biology that are shedding new light by untangling how subtle changes in conserved genes, pathways, and epigenetic factors influence organismal senescence and associated declines. Biogerontology is a complex and rapidly growing field aimed at elucidating genetic modifications that extend lifespan and healthspan. This review explores gerontogenes, genes influencing lifespan and healthspan across species. Though subtle differences exist, long-lived individuals such as centenarians demonstrate extended healthspans, and numerous studies confirm the heritability of longevity/healthspan genes. Importantly, genes and gerontogenes are directly and indirectly involved in DNA repair, insulin/IGF-1 and mTOR signaling pathways, long non-coding RNAs, sirtuins, and heat shock proteins. The complex interactions between genetics and epigenetics are teased apart. While more research into optimizing healthspan is needed, conserved gerontogenes offer synergistic potential to forestall aging and age-related diseases. Understanding complex longevity genetics brings closer the goal of extending not only lifespan but quality years of life. The primary aim of human Biogerontology is to enhance lifespan and healthspan, but the question remains: are current genetic modifications effectively promoting healthy aging? This article collates the advancements in gerontogenes that enhance lifespan and improve healthspan alongside their potential challenges.
Collapse
Affiliation(s)
| | - Enock D Mensah
- Chemistry, Virginia Polytechnic Institute and State University, Blacksburg, USA
| | - Kwabena Dabie
- Chemistry and Chemical Biology, University of New Mexico, Albuquerque, USA
| | - Caleb Mensah
- Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Blacksburg, USA
| | | | - Jonathan Essuman
- School of Molecular Sciences, Arizona State University, Tempe, USA
| |
Collapse
|
23
|
Liu Y, Ouyang Y, Yu L, Wang P, Peng Z, Liu H, Zhao S, Wang H, Zhou Z, Deng Y, Liu Y, Xie J. Novel approach for enhancing skin allograft survival by bioadhesive nanoparticles loaded with rapamycin. Int J Pharm 2024; 651:123742. [PMID: 38151102 DOI: 10.1016/j.ijpharm.2023.123742] [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: 09/05/2023] [Revised: 12/13/2023] [Accepted: 12/22/2023] [Indexed: 12/29/2023]
Abstract
Skin graft rejection is a significant challenge in skin allografts for skin defects, particularly in extensive burn injury patients when autografts are insufficient. Enhancing the survival duration of allogeneic skin grafts can improve the success rate of subsequent autologous skin grafting, thereby promoting the therapeutic efficacy for wound healing. Rapamycin (Rapa), a potent immunosuppressant with favorable efficacy in organ transplantation, is limited by its systemic administration-associated toxicity and side effects. Therefore, addressing the short survival time of allogeneic skin grafts and minimizing the toxicity related to systemic application of immunosuppressive agents is an urgent requirement. Here, we present a topical formulation based on bioadhesive poly (lactic acid)-hyperbranched polyglycerol nanoparticles (BNPs) with surface-modified encapsulation of Rapamycin (Rapa/BNPs), applied for local immunosuppression in a murine model of allogeneic skin grafts. Our Rapa/BNPs significantly prolong nanoparticle retention, reduce infiltration of T lymphocytes and macrophages, decrease the level of pro-inflammatory cytokines and ultimately extend skin allograft survival with little systemic toxicity compared to free Rapa or Rapamycin-loaded non-bioadhesive nanoparticles (Rapa/NNPs) administration. In conclusion, Rapa/BNPs effectively deliver local immunosuppression and demonstrate potential for enhancing skin allograft survival while minimizing localized inflammation, thus potentially increasing patient survival rates for various types of skin defects.
Collapse
Affiliation(s)
- Yiling Liu
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yaqi Ouyang
- Department of School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China
| | - Liu Yu
- Department of School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China
| | - Peng Wang
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Zhangwen Peng
- Department of School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China
| | - Hengdeng Liu
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Shixin Zhao
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Hanwen Wang
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ziheng Zhou
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yang Deng
- Department of School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China.
| | - Yang Liu
- Department of School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China; Department of School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No.66, Gongchang Road, Shenzhen, 518107, China.
| | - Julin Xie
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No.58, Zhongshan 2nd Road, Guangzhou, 510080, China.
| |
Collapse
|
24
|
Hong J, Wang X, Jin H, Chen Y, Jiang Y, Du K, Chen D, Zheng S, Cao L. Environment relevant exposure of perfluorooctanoic acid accelerates the growth of hepatocellular carcinoma cells through mammalian target of rapamycin (mTOR) signal pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 341:122910. [PMID: 37967710 DOI: 10.1016/j.envpol.2023.122910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/25/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
Perfluorooctanoic acid (PFOA), a synthetic alkyl chain fluorinated compound, has emerged as a persistent organic pollutant of grave concern, casting a shadow over both ecological integrity and humans. Its insidious presence raises alarms due to its capacity to bioaccumulate within the human liver, potentially paving the treacherous path toward liver cancer. Yet, the intricate mechanisms underpinning PFOA's role in promoting the growth of hepatocellular carcinoma (HCC) remain shrouded in ambiguity. Here, we determined the proliferation and transcription changes of HCC after PFOA exposure through integrated experiments including cell culture, nude mice tests, and colony-forming assays. Based on our findings, PFOA effectively promotes the proliferation of HCC cells within the experimental range of concentrations, both in vivo and in vitro. The proliferation efficiency of HCC cells was observed to increase by approximately 10% due to overexposure to PFOA. Additionally, the cancer weight of tumor-bearing nude mice increased by 87.0% (p < 0.05). We systematically evaluated the effects of PFOA on HCC cells and found that PFOA's exposure can selectively activate the PI3K/AKT/mTOR/4E-BP1 signaling pathway, thereby playing a pro-cancer effect on HCC cells Confirmation echoed through western blot assays and inhibitor combination analyses. These insights summon a response to PFOA's dual nature as both an environmental threat and a promoter of liver cancer. Our work illuminates the obscured domain of PFOA-induced hepatoxicity, shedding light on its ties to hepatocellular carcinoma progression.
Collapse
Affiliation(s)
- Jiawei Hong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, PR China
| | - Xiaoyan Wang
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China
| | - Hangbiao Jin
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang, 310032, PR China
| | - Yuanchen Chen
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou, Zhejiang, 310032, PR China
| | - Yifan Jiang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, PR China
| | - Keyi Du
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, PR China
| | - Diyu Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, PR China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, PR China
| | - Linping Cao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, PR China; Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, PR China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, PR China.
| |
Collapse
|
25
|
Cullen ER, Safari M, Mittelstadt I, Weston MC. Hyperactivity of mTORC1 and mTORC2-dependent signaling mediate epilepsy downstream of somatic PTEN loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.18.553856. [PMID: 37645923 PMCID: PMC10462128 DOI: 10.1101/2023.08.18.553856] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Gene variants that hyperactivate PI3K-mTOR signaling in the brain lead to epilepsy and cortical malformations in humans. Some gene variants associated with these pathologies only hyperactivate mTORC1, but others, such as PTEN, PIK3CA, and AKT, hyperactivate both mTORC1- and mTORC2-dependent signaling. Previous work established a key role for mTORC1 hyperactivity in mTORopathies, however, whether mTORC2 hyperactivity contributes is not clear. To test this, we inactivated mTORC1 and/or mTORC2 downstream of early Pten deletion in a new model of somatic Pten loss-of-function (LOF) in the cortex and hippocampus. Spontaneous seizures and epileptiform activity persisted despite mTORC1 or mTORC2 inactivation alone, but inactivating both mTORC1 and mTORC2 simultaneously normalized brain activity. These results suggest that hyperactivity of both mTORC1 and mTORC2 can cause epilepsy, and that targeted therapies should aim to reduce activity of both complexes.
Collapse
Affiliation(s)
- Erin R. Cullen
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington VT, 05405, USA
| | - Mona Safari
- Fralin Biomedical Research Institute at VTC, Center for Neurobiology Research, Roanoke VA, 24016, USA
| | - Isabelle Mittelstadt
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington VT, 05405, USA
| | - Matthew C. Weston
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington VT, 05405, USA
- Fralin Biomedical Research Institute at VTC, Center for Neurobiology Research, Roanoke VA, 24016, USA
- School of Neuroscience, Virginia Polytechnic and State University, Blacksburg VA, 24060, USA
| |
Collapse
|
26
|
Pérez-Martínez L, Romero L, Verdugo-Sivianes EM, Muñoz-Galván S, Rubio-Mediavilla S, Amiama-Roig A, Carnero A, Blanco JR. Role of maraviroc and/or rapamycin in the liver of IL10 KO mice with frailty syndrome. PLoS One 2024; 19:e0286201. [PMID: 38198476 PMCID: PMC10781157 DOI: 10.1371/journal.pone.0286201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/10/2023] [Indexed: 01/12/2024] Open
Abstract
Cellular senescence and low-grade inflammation favor the acceleration of aging. The liver is an essential metabolic organ because changes related to its function are related to age-related diseases. The objective of this study was to evaluate the effects of maraviroc (MVC) and/or rapamycin (RAPA) on liver tissue in an experimental model of frailty syndrome in mice, since MVC and RAPA are two molecules able to decrease CCR5 expression, which is overexpressed in patients with frailty. Methods: Eighty male homozygous IL10KO mice were randomly assigned to one of 4 groups (n = 20): i) IL10KO group; ii) MVC group, iii) RAPA group, and iv) MVC-RAPA group. Liver samples were analyzed. Gene expression quantification and western blotting were also performed. The proinflammatory cytokines IL-6 and IL-18 were decreased in MVC and MVC/RAPA groups, IL-12 was decreased in RAPA and MVC/RAPA groups and TNF-α was decreased in all therapeutic groups. P21 was decreased in RAPA and MVC/RAPA groups, Galactosidase beta-1, was also significantly reduced in all therapeutic groups, as were NF-kB1, NF-kB2 and STAT3. In all groups, mTOR and CCL5 were significantly reduced. CCR5 expression was decreased in the MVC and MVC/RAPA groups. Conclusion: MVC and RAPA may protect against some factors involved in liver aging. More studies will be necessary to verify their clinical applications.
Collapse
Affiliation(s)
| | - Lourdes Romero
- Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain
| | - Eva M. Verdugo-Sivianes
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Sevilla, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Muñoz-Galván
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Sevilla, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Ana Amiama-Roig
- Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Sevilla, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - José-Ramón Blanco
- Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain
- Servicio de Enfermedades Infecciosas, Hospital Universitario San Pedro, Logroño, Spain
| |
Collapse
|
27
|
Sonis ST, Villa A. A New Hypothesis Describing the Pathogenesis of Oral Mucosal Injury Associated with the Mammalian Target of Rapamycin (mTOR) Inhibitors. Cancers (Basel) 2023; 16:68. [PMID: 38201496 PMCID: PMC10777973 DOI: 10.3390/cancers16010068] [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: 11/27/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
It has been 24 years since rapamycin (sirolimus) was approved to mitigate solid organ transplant rejection and 16 years since mTOR (mammalian/mechanistic target of rapamycin) inhibitors reached patients as a cancer therapy. While the clinical benefits of mTOR inhibitors (mTORi) are robust, so too are their toxicities. Among the most common issues is the development of ulcers of the oral mucosa (mTOR-inhibitor associated stomatitis; mIAS). These lesions are distinct from those of other anti-cancer agents, occur with regularity, and impact patient outcomes. mIAS' pathogenesis has been the subject of speculation, and its similar presentation to recurrent aphthous stomatitis (RAS) has led to the hypothesis that it might serve as a surrogate to better understand RAS. Based on a review of the literature, the current manuscript provides a hypothesis regarding the mechanisms by which mTORis uniquely initiate mucosal injury and an explanation for the observation that steroids (also an immunosuppressive) are effective in its treatment through a non-immunologic mechanism. Unexplained unique features of mIAS are discussed in this review in the context of future investigation.
Collapse
Affiliation(s)
- Stephen T. Sonis
- Divisions of Oral Medicine and Dentistry, Brigham and Women’s Hospital and the Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02114, USA
- Biomodels, LLC, Waltham, MA 02451, USA
| | - Alessandro Villa
- Oral Medicine, Oral Oncology and Dentistry, Miami Cancer Institute, Miami, FL 33176, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
28
|
李 彬, 汪 越, 侯 凤, 杜 家, 童 旭. [Rapamycin enhances inhibitory effect of RSL3 on proliferation, invasion and migration of testicular cancer I-10 cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:2145-2151. [PMID: 38189403 PMCID: PMC10774114 DOI: 10.12122/j.issn.1673-4254.2023.12.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Indexed: 01/09/2024]
Abstract
OBJECTIVE To investigate the effect of rapamycin for enhancing the inhibitory effect of RSL3 on proliferation, invasion and migration of testicular cancer I-10 cells in vitro. METHODS I-10 cells treated with 0-16 μmol/L RSL3 (Type Ⅱ) either alone or in combination with 16 μmol/L rapamycin (RAPA) were examined for changes in proliferation using MTT assay and colonyforming assay, and the changes in cell migration and invasion abilities were detected with wounding-healing assay and Transwell assay. The changes in the levels of lipid reactive oxygen species in the treated cells were detected using flow cytometry. GSH and MDA contents in the cells were detected using commercial detection kits, and GPX4 protein expression level was determined with Western blotting. RESULTS The cytotoxic effect of RSL3 increased dose-dependently in I-10 cells, and the combined treatment with rapamycin further enhanced its cytotoxicity. Treatment of I-10 cells with RSL3 alone significantly decreased cell colony numbers (P < 0.05), wounding-healing rates (P < 0.01), and invasion and migration cell numbers (P < 0.05), increased lipid reactive oxygen species level and MDA content (P < 0.05), and lowered GSH content and expression level of GPX4 protein in the cells (P < 0.01). The inhibitory effects of RSL3 were significantly enhanced by co-treatment of the cells with rapamycin (P < 0.05 or 0.01). CONCLUSION Rapamycin enhances the inhibitory effect of RSL3 on proliferation, invasion and migration of I-10 cells by enhancing RSL3-mediated cell ferroptosis.
Collapse
Affiliation(s)
- 彬 李
- />蚌埠医学院药学院/安徽省生化药物工程技术研究中心,安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Research Center for Biochemical Pharmaceuticals, Bengbu 233030, China
| | - 越 汪
- />蚌埠医学院药学院/安徽省生化药物工程技术研究中心,安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Research Center for Biochemical Pharmaceuticals, Bengbu 233030, China
| | - 凤伟 侯
- />蚌埠医学院药学院/安徽省生化药物工程技术研究中心,安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Research Center for Biochemical Pharmaceuticals, Bengbu 233030, China
| | - 家如 杜
- />蚌埠医学院药学院/安徽省生化药物工程技术研究中心,安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Research Center for Biochemical Pharmaceuticals, Bengbu 233030, China
| | - 旭辉 童
- />蚌埠医学院药学院/安徽省生化药物工程技术研究中心,安徽 蚌埠 233030School of Pharmacy, Bengbu Medical College, Anhui Provincial Engineering Research Center for Biochemical Pharmaceuticals, Bengbu 233030, China
| |
Collapse
|
29
|
Elliehausen CJ, Anderson RM, Diffee GM, Rhoads TW, Lamming DW, Hornberger TA, Konopka AR. Geroprotector drugs and exercise: friends or foes on healthy longevity? BMC Biol 2023; 21:287. [PMID: 38066609 PMCID: PMC10709984 DOI: 10.1186/s12915-023-01779-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Physical activity and several pharmacological approaches individually combat age-associated conditions and extend healthy longevity in model systems. It is tantalizing to extrapolate that combining geroprotector drugs with exercise could extend healthy longevity beyond any individual treatment. However, the current dogma suggests that taking leading geroprotector drugs on the same day as exercise may limit several health benefits. Here, we review leading candidate geroprotector drugs and their interactions with exercise and highlight salient gaps in knowledge that need to be addressed to identify if geroprotector drugs can have a harmonious relationship with exercise.
Collapse
Affiliation(s)
- Christian J Elliehausen
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research, Education, and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Rozalyn M Anderson
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research, Education, and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Gary M Diffee
- Department of Kinesiology, University of Wisconsin-Madison, Madison, WI, USA
| | - Timothy W Rhoads
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W Lamming
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Troy A Hornberger
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam R Konopka
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- Geriatric Research, Education, and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
30
|
Goul C, Peruzzo R, Zoncu R. The molecular basis of nutrient sensing and signalling by mTORC1 in metabolism regulation and disease. Nat Rev Mol Cell Biol 2023; 24:857-875. [PMID: 37612414 DOI: 10.1038/s41580-023-00641-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2023] [Indexed: 08/25/2023]
Abstract
The Ser/Thr kinase mechanistic target of rapamycin (mTOR) is a central regulator of cellular metabolism. As part of mTOR complex 1 (mTORC1), mTOR integrates signals such as the levels of nutrients, growth factors, energy sources and oxygen, and triggers responses that either boost anabolism or suppress catabolism. mTORC1 signalling has wide-ranging consequences for the growth and homeostasis of key tissues and organs, and its dysregulated activity promotes cancer, type 2 diabetes, neurodegeneration and other age-related disorders. How mTORC1 integrates numerous upstream cues and translates them into specific downstream responses is an outstanding question with major implications for our understanding of physiology and disease mechanisms. In this Review, we discuss recent structural and functional insights into the molecular architecture of mTORC1 and its lysosomal partners, which have greatly increased our mechanistic understanding of nutrient-dependent mTORC1 regulation. We also discuss the emerging involvement of aberrant nutrient-mTORC1 signalling in multiple diseases.
Collapse
Affiliation(s)
- Claire Goul
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Roberta Peruzzo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|
31
|
Li X, Peng Z, An K, Xue M, Wang Z, Xia J, Qi Z, Shu X. Temsirolimus is a promising immunomodulatory agent for enhanced transplantation outcomes. Transpl Immunol 2023; 81:101952. [PMID: 37918580 DOI: 10.1016/j.trim.2023.101952] [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: 08/02/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Identifying effective immunosuppressive strategies is critical for addressing immunological rejection following organ transplantation. This study explores the potential immunosuppressive effects and mechanisms of temsirolimus, a rapamycin derivative, in organ transplantation. METHODS A mouse cardiac allograft model was established using a cervical cannula technique with BALB/c donors and C57BL/6 recipients. Mice were administered temsirolimus intragastrically and graft survival was evaluated. Histological staining was used to assess pathological changes. The BrdU assay was used to measure splenic T cell proliferation. Flow cytometry was used to quantify regulatory T cells (Tregs), CD4+ T cells, and CD8+ T cells. ELISA and qPCR assays were used to determine Foxp3, IL-4, IFN-γ, and TGF-β expression. RESULTS Temsirolimus displayed potent immunosuppressive effects at 20 mg/kg/day, significantly inhibiting T cell proliferation (84.6%, P < 0.0001) and prolonging graft survival (median 49 days vs. 8.5 days in controls, P < 0.0001). However, median survival decreased to 34.5 days upon withdrawal. Temsirolimus also reduced splenic CD4+ and CD8+ T cells (2.85% and 2.92%, P < 0.001) and antibody levels (IgM, IgG1, IgG2) by 11.85-29.09% (P < 0.0001) and increased Tregs, Foxp3, IL-4 (P < 0.01), and TGF-β (P < 0.05), while decreasing IFN-γ (P < 0.001). CONCLUSIONS Temsirolimus exhibited potent immunosuppressive effects, emerging as a strong candidate to mitigate organ transplant rejection.
Collapse
Affiliation(s)
- Xianguo Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zuojie Peng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke An
- Department of Physiology, Xuzhou Medical University, Xuzhou 221009, China
| | - Mengjiao Xue
- Division of Ophthalmology and Vision Science, Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhenzhen Wang
- Department of Pharmacy, Zhoukou Central Hospital, Zhoukou 466000, China
| | - Junjie Xia
- Organ Transplantation Institute, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen 361100, China.
| | - Zhongquan Qi
- Medical College of Guangxi University, Guangxi University, Nanning 530004, China.
| | - Xiaogang Shu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
32
|
Lopez JJD, Gaza JT, Nellas RB. The role of glycerol-water mixtures in the stability of FKBP12-rapalog-FRB complexes. J Mol Graph Model 2023; 124:108556. [PMID: 37423019 DOI: 10.1016/j.jmgm.2023.108556] [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: 11/12/2022] [Revised: 04/27/2023] [Accepted: 06/16/2023] [Indexed: 07/11/2023]
Abstract
The thermodynamic and biophysical implications of the introduction of a co-solvent during protein-ligand binding remain elusive. Using ternary complexes of 12-kDa FK506 binding protein (FKBP12), FKBP-rapamycin binding (FRB) domain of the mammalian/mechanistic target of rapamycin (mTOR) kinase, and rapamycin analogs (rapalogs) in glycerol-water mixtures, the influence of solvent composition on ligand binding dynamics was explored. The pharmaceutical potential of rapalogs and the utility of glycerol as a co-solvent in drug delivery applications were critical in deciding the system to be studied. Consolidation of existing studies on rapamycin modification was first performed to strategically design a new rapalog called T1. The results from 100-ns dual-boost Gaussian accelerated molecular dynamics simulations showed that protein stability was induced in the presence of glycerol. Reweighting of the trajectories revealed that the glycerol-rich solvent system lowers the energy barrier in the conformational space of the protein while also preserving native contacts between the ligand and the residues in the binding site. Calculated binding free energies using MM/GBSA also showed that electrostatic energy and polar contribution of solvation energy are heavily influenced by the changes in solvation. Glycerol molecules are preferentially excluded through electrostatic interactions from the solvation shell which induce complex stability as seen in existing experiments. Hence, using glycerol as a co-solvent in rapamycin delivery has a significant role in maintaining stability. In addition, compound T1 is a potential mTORC1-selective inhibitor with strong affinity for the FKBP12-FRB complex. This study aims to provide insights on the design of new rapalogs, and the applicability of glycerol as co-solvent for FKBP12-rapalog-FRB complexes.
Collapse
Affiliation(s)
- Joshua Jener D Lopez
- Institute of Chemistry, College of Science, University of the Philippines Diliman, Quezon City, 1101, Philippines
| | - Jokent T Gaza
- Institute of Chemistry, College of Science, University of the Philippines Diliman, Quezon City, 1101, Philippines
| | - Ricky B Nellas
- Institute of Chemistry, College of Science, University of the Philippines Diliman, Quezon City, 1101, Philippines.
| |
Collapse
|
33
|
Wang H, Yang R, Wang Z, Cao L, Kong D, Sun Q, Yoshida S, Ren J, Chen T, Duan J, Lu J, Shen Z, Zheng H. Metronomic capecitabine with rapamycin exerts an immunosuppressive effect by inducing ferroptosis of CD4 + T cells after liver transplantation in rat. Int Immunopharmacol 2023; 124:110810. [PMID: 37625370 DOI: 10.1016/j.intimp.2023.110810] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/02/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023]
Abstract
Liver transplantation is one of the most effective treatments for hepatocellular carcinoma (HCC). The balance between inhibiting immune rejection and preventing tumor recurrence after liver transplantation is the key to determining the long-term prognosis of patients with HCC after liver transplantation. In our previous study, we found that capecitabine (CAP), an effective drug for the treatment of HCC, could exert an immunosuppressive effect after liver transplantation by inducing T cell ferroptosis. Recent studies have shown that ferroptosis is highly associated with autophagy. In this study, we confirmed that the autophagy inducer rapamycin (RAPA) combined with metronomic capecitabine (mCAP) inhibits glutathione peroxidase 4 (GPX4) and promotes ferroptosis in CD4+ T cells to exert immunosuppressive effects after rat liver transplantation. Compared with RAPA or mCAP alone, the combination of RAPA and mCAP could adequately reduce liver injury in rats with acute rejection after transplantation. The CD4+ T cell counts in peripheral blood, spleen, and transplanted liver of recipient rats significantly decreased, and the oxidative stress level and ferrous ion concentration of CD4+ T cells significantly increased in the combination group. In vitro, the combination of drugs significantly promoted autophagy, decreased GPX4 protein expression, and induced ferroptosis in CD4+ T cells. In conclusion, the autophagy inducer RAPA improved the mCAP-induced ferroptosis in CD4+ T cells. Our results support the concept of ferroptosis as an autophagy-dependent cell death and suggest that the combination of ferroptosis inducers and autophagy inducers is a new research direction for improving immunosuppressive regimens after liver transplantation.
Collapse
Affiliation(s)
- Hao Wang
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Ruining Yang
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Zhenglu Wang
- Organ Transplant Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Lei Cao
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Dejun Kong
- School of Medicine, Nankai University, Tianjin, China
| | - Qian Sun
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Sei Yoshida
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China
| | - Jiashu Ren
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Tao Chen
- School of Medicine, Nankai University, Tianjin, China
| | - Jinliang Duan
- School of Medicine, Nankai University, Tianjin, China
| | - Jianing Lu
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Zhongyang Shen
- Organ Transplant Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China; National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin, China
| | - Hong Zheng
- Organ Transplant Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China; National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin, China.
| |
Collapse
|
34
|
Yepuri G, Ramirez LM, Theophall GG, Reverdatto SV, Quadri N, Hasan SN, Bu L, Thiagarajan D, Wilson R, Díez RL, Gugger PF, Mangar K, Narula N, Katz SD, Zhou B, Li H, Stotland AB, Gottlieb RA, Schmidt AM, Shekhtman A, Ramasamy R. DIAPH1-MFN2 interaction regulates mitochondria-SR/ER contact and modulates ischemic/hypoxic stress. Nat Commun 2023; 14:6900. [PMID: 37903764 PMCID: PMC10616211 DOI: 10.1038/s41467-023-42521-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 10/13/2023] [Indexed: 11/01/2023] Open
Abstract
Inter-organelle contact and communication between mitochondria and sarco/endoplasmic reticulum (SR/ER) maintain cellular homeostasis and are profoundly disturbed during tissue ischemia. We tested the hypothesis that the formin Diaphanous-1 (DIAPH1), which regulates actin dynamics, signal transduction and metabolic functions, contributes to these processes. We demonstrate that DIAPH1 interacts directly with Mitofusin-2 (MFN2) to shorten mitochondria-SR/ER distance, thereby enhancing mitochondria-ER contact in cells including cardiomyocytes, endothelial cells and macrophages. Solution structure studies affirm the interaction between the Diaphanous Inhibitory Domain and the cytosolic GTPase domain of MFN2. In male rodent and human cardiomyocytes, DIAPH1-MFN2 interaction regulates mitochondrial turnover, mitophagy, and oxidative stress. Introduction of synthetic linker construct, which shorten the mitochondria-SR/ER distance, mitigated the molecular and functional benefits of DIAPH1 silencing in ischemia. This work establishes fundamental roles for DIAPH1-MFN2 interaction in the regulation of mitochondria-SR/ER contact networks. We propose that targeting pathways that regulate DIAPH1-MFN2 interactions may facilitate recovery from tissue ischemia.
Collapse
Affiliation(s)
- Gautham Yepuri
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Lisa M Ramirez
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, 12222, USA
| | - Gregory G Theophall
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, 12222, USA
| | - Sergei V Reverdatto
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, 12222, USA
| | - Nosirudeen Quadri
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Syed Nurul Hasan
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Lei Bu
- Department of Medicine, Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Devi Thiagarajan
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Robin Wilson
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Raquel López Díez
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Paul F Gugger
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Kaamashri Mangar
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Navneet Narula
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Stuart D Katz
- Department of Medicine, Leon H. Charney Division of Cardiology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Boyan Zhou
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Huilin Li
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Aleksandr B Stotland
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Roberta A Gottlieb
- Department of Biomedical Sciences, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA
| | - Alexander Shekhtman
- Department of Chemistry, University of Albany, State University of New York, Albany, NY, 12222, USA
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York, 10016, USA.
| |
Collapse
|
35
|
Konopka AR, Lamming DW. Blazing a trail for the clinical use of rapamycin as a geroprotecTOR. GeroScience 2023; 45:2769-2783. [PMID: 37801202 PMCID: PMC10643772 DOI: 10.1007/s11357-023-00935-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/04/2023] [Indexed: 10/07/2023] Open
Abstract
Treatment with rapamycin, an inhibitor of the mechanistic Target Of Rapamycin Complex One (mTORC1) protein kinase, has been repeatedly demonstrated to extend lifespan and prevent or delay age-related diseases in diverse model systems. Concerns over the risk of potentially serious side effects in humans, including immunosuppression and metabolic disruptions, have cautiously limited the translation of rapamycin and its analogs as a treatment for aging associated conditions. During the last decade, we and others have developed a working model that suggests that while inhibition of mTORC1 promotes healthy aging, many of the negative side effects of rapamycin are associated with "off-target" inhibition of a second mTOR complex, mTORC2. Differences in the kinetics and molecular mechanisms by which rapamycin inhibits mTORC1 and mTORC2 suggest that a therapeutic window for rapamycin could be exploited using intermittent dosing schedules or alternative rapalogs that may enable more selective inhibition of mTORC1. However, the optimal dosing schedules and the long-term efficacy of such interventions in humans are unknown. Here, we highlight ongoing or upcoming clinical trials that will address outstanding questions regarding the safety, pharmacokinetics, pharmacodynamics, and efficacy of rapamycin and rapalogs on several clinically oriented outcomes. Results from these early phase studies will help guide the design of phase 3 clinical trials to determine whether rapamycin can be used safely to inhibit mTORC1 for the treatment and prevention of age-related diseases in humans.
Collapse
Affiliation(s)
- Adam R Konopka
- Division of Geriatrics, Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA.
- Division of Geriatrics and Gerontology, Department of Medicine, Geriatric Research Education and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, University of Wisconsin-Madison, 2500 Overlook Terrace, Madison, WI, 53705, USA.
| | - Dudley W Lamming
- William S. Middleton Memorial Veterans Hospital, Madison, WI, 53705, USA
- Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
| |
Collapse
|
36
|
Choi HK, Park SH, Lee J, Hwang JT. Review of Patents for Anticancer Agents Targeting Adenosine Monophosphate-Activated Protein Kinase. J Med Food 2023; 26:605-615. [PMID: 37590001 DOI: 10.1089/jmf.2023.k.0026] [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] [Indexed: 08/18/2023] Open
Abstract
Cancer, caused by abnormal and excessive cellular proliferation, can invade and destroy surrounding tissues and organs through the spreading of cancer cells. A general strategy for developing anticancer agents is to identify biomarkers that, if targeted, can produce a robust cytotoxic effect with minimal side effects. Cell-cycle regulators, checkpoint regulatory genes, and apoptosis-related genes are well-known biomarkers that inhibit cancer cell proliferation. Several compounds that target such biomarkers have been patented and more are being developed as novel therapies. Recent additions to this list include anticancer drugs that target signaling pathway proteins, such as 5' adenosine monophosphate-activated protein kinase (AMPK), which plays a vital role in cancer and normal cell metabolism. Herein, we have reviewed recent patents related to AMPK-targeting anticancer drugs and discussed the mechanisms of action of these drugs. We conclude that these recently published patents include several attractive compounds and methods for targeting AMPK. Further research and clinical trials are required to elucidate the comprehensive role of AMPK in cancer cell metabolism, identify its associated signal transduction systems, and develop novel activators that may find applications in cancer therapy. Clinical Trial Registration number: NCT01904123.
Collapse
Affiliation(s)
- Hyo-Kyoung Choi
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| | - Soo-Hyun Park
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| | - Jangho Lee
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| | - Jin-Taek Hwang
- Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| |
Collapse
|
37
|
Berquez M, Chen Z, Festa BP, Krohn P, Keller SA, Parolo S, Korzinkin M, Gaponova A, Laczko E, Domenici E, Devuyst O, Luciani A. Lysosomal cystine export regulates mTORC1 signaling to guide kidney epithelial cell fate specialization. Nat Commun 2023; 14:3994. [PMID: 37452023 PMCID: PMC10349091 DOI: 10.1038/s41467-023-39261-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 06/06/2023] [Indexed: 07/18/2023] Open
Abstract
Differentiation is critical for cell fate decisions, but the signals involved remain unclear. The kidney proximal tubule (PT) cells reabsorb disulphide-rich proteins through endocytosis, generating cystine via lysosomal proteolysis. Here we report that defective cystine mobilization from lysosomes through cystinosin (CTNS), which is mutated in cystinosis, diverts PT cells towards growth and proliferation, disrupting their functions. Mechanistically, cystine storage stimulates Ragulator-Rag GTPase-dependent recruitment of mechanistic target of rapamycin complex 1 (mTORC1) and its constitutive activation. Re-introduction of CTNS restores nutrient-dependent regulation of mTORC1 in knockout cells, whereas cell-permeant analogues of L-cystine, accumulating within lysosomes, render wild-type cells resistant to nutrient withdrawal. Therapeutic mTORC1 inhibition corrects lysosome and differentiation downstream of cystine storage, and phenotypes in preclinical models of cystinosis. Thus, cystine serves as a lysosomal signal that tailors mTORC1 and metabolism to direct epithelial cell fate decisions. These results identify mechanisms and therapeutic targets for dysregulated homeostasis in cystinosis.
Collapse
Affiliation(s)
- Marine Berquez
- Institute of Physiology, University of Zurich, 8057, Zurich, Switzerland
| | - Zhiyong Chen
- Institute of Physiology, University of Zurich, 8057, Zurich, Switzerland
| | | | - Patrick Krohn
- Institute of Physiology, University of Zurich, 8057, Zurich, Switzerland
| | | | - Silvia Parolo
- Fondazione The Microsoft Research University of Trento-Centre for Computational and Systems Biology (COSBI), 38068, Rovereto, Italy
| | - Mikhail Korzinkin
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Anna Gaponova
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Endre Laczko
- Functional Genomics Center Zurich, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Enrico Domenici
- Fondazione The Microsoft Research University of Trento-Centre for Computational and Systems Biology (COSBI), 38068, Rovereto, Italy
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, 8057, Zurich, Switzerland.
- Institute for Rare Diseases, UCLouvain Medical School, 1200, Brussels, Belgium.
| | - Alessandro Luciani
- Institute of Physiology, University of Zurich, 8057, Zurich, Switzerland.
| |
Collapse
|
38
|
Mannick JB, Lamming DW. Targeting the biology of aging with mTOR inhibitors. NATURE AGING 2023; 3:642-660. [PMID: 37142830 PMCID: PMC10330278 DOI: 10.1038/s43587-023-00416-y] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/07/2023] [Indexed: 05/06/2023]
Abstract
Inhibition of the protein kinase mechanistic target of rapamycin (mTOR) with the Food and Drug Administration (FDA)-approved therapeutic rapamycin promotes health and longevity in diverse model organisms. More recently, specific inhibition of mTORC1 to treat aging-related conditions has become the goal of basic and translational scientists, clinicians and biotechnology companies. Here, we review the effects of rapamycin on the longevity and survival of both wild-type mice and mouse models of human diseases. We discuss recent clinical trials that have explored whether existing mTOR inhibitors can safely prevent, delay or treat multiple diseases of aging. Finally, we discuss how new molecules may provide routes to the safer and more selective inhibition of mTOR complex 1 (mTORC1) in the decade ahead. We conclude by discussing what work remains to be done and the questions that will need to be addressed to make mTOR inhibitors part of the standard of care for diseases of aging.
Collapse
Affiliation(s)
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
39
|
Qi C, Bujaroski RS, Baell J, Zheng X. Kinases in cerebral cavernous malformations: Pathogenesis and therapeutic targets. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119488. [PMID: 37209718 DOI: 10.1016/j.bbamcr.2023.119488] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/03/2023] [Accepted: 05/11/2023] [Indexed: 05/22/2023]
Abstract
Cerebral cavernous malformations (CCMs) are low-flow, hemorrhagic vascular lesions of the central nervous system of genetic origin, which can cause stroke-like symptoms and seizures. From the identification of CCM1, CCM2 and CCM3 as genes related to disease progression, molecular and cellular mechanisms for CCM pathogenesis have been established and the search for potential drugs to target CCM has begun. Broadly speaking, kinases are the major group signaling in CCM pathogenesis. These include the MEKK3/MEK5/ERK5 cascade, Rho/Rock signaling, CCM3/GCKIII signaling, PI3K/mTOR signaling, and others. Since the discovery of Rho/Rock in CCM pathogenesis, inhibitors for Rho signaling and subsequently other components in CCM signaling were discovered and applied in preclinical and clinical trials to ameliorate CCM progression. This review discusses the general aspects of CCM disease, kinase-mediated signaling in CCM pathogenesis and the current state of potential treatment options for CCM. It is suggested that kinase target drug development in the context of CCM might facilitate and meet the unmet requirement - a non-surgical option for CCM disease.
Collapse
Affiliation(s)
- Chunxiao Qi
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, China
| | - Richard Sean Bujaroski
- Medicinal Chemistry Theme, Monash Institute of Pharmaceutical Sciences, Australian Translational Medicinal Chemistry Facility (ATMCF), Monash University, Parkville, Victoria, Australia
| | - Jonathan Baell
- School of Pharmaceutical Sciences, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing 211816, China
| | - Xiangjian Zheng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, China.
| |
Collapse
|
40
|
Parate S, Kumar V, Hong JC, Lee KW. Investigation of Macrocyclic mTOR Modulators of Rapamycin Binding Site via Pharmacoinformatics Approaches. Comput Biol Chem 2023; 104:107875. [PMID: 37148678 DOI: 10.1016/j.compbiolchem.2023.107875] [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: 07/25/2022] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/08/2023]
Abstract
The PI3K/Akt/mTOR is an essential intracellular signaling pathway in which the serine/threonine mTOR kinase portrays a major role in cell growth, proliferation and survival. The mTOR kinase is frequently dysregulated in a broad spectrum of cancers, thus making it a potential target. Rapamycin and its analogs (rapalogs) allosterically inhibit mTOR, thereby dodging the deleterious effects prompted by ATP-competitive mTOR inhibitors. However, the available mTOR allosteric site inhibitors exhibit low oral bioavailability and suboptimal solubility. Bearing in mind this narrow therapeutic window of the current allosteric mTOR inhibitors, an in silico study was designed in search of new macrocyclic inhibitors. The macrocycles from the ChemBridge database (12,677 molecules) were filtered for their drug-likeness properties and the procured compounds were subjected for molecular docking within the binding cleft between FKBP25 and FRB domains of mTOR. The docking analysis resulted with 15 macrocycles displaying higher scores than the selective mTOR allosteric site inhibitor, DL001. The docked complexes were refined by subsequent molecular dynamics simulations for a period of 100 ns. Successive binding free energy computation revealed a total of 7 macrocyclic compounds (HITS) demonstrating better binding affinity than DL001, towards mTOR. The consequent assessment of pharmacokinetic properties resulted in HITS with similar or better properties than the selective inhibitor, DL001. The HITS from this investigation could act as effective mTOR allosteric site inhibitors and serve as macrocyclic scaffolds for developing compounds targeting the dysregulated mTOR.
Collapse
Affiliation(s)
- Shraddha Parate
- Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Division of Applied Life Science, Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, South Korea; Department of Chemistry and Molecular Biology, University of Gothenburg, 405 30 Göteborg, Sweden.
| | - Vikas Kumar
- Department of Bio & Medical Big Data (BK4 Program), Division of Life Sciences, Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, South Korea
| | - Jong Chan Hong
- Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Division of Applied Life Science, Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, South Korea.
| | - Keun Woo Lee
- Department of Bio & Medical Big Data (BK4 Program), Division of Life Sciences, Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, South Korea.
| |
Collapse
|
41
|
Tian J, Wang J, Xu H, Zou B, Chen W, Liu Y, Chen J, Zhang R. Nanoscale metal-organic framework delivers rapamycin to induce tissue immunogenic cell death and potentiates cancer immunotherapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 50:102678. [PMID: 37044194 DOI: 10.1016/j.nano.2023.102678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/21/2023] [Accepted: 04/02/2023] [Indexed: 04/14/2023]
Abstract
Rapamycin has great potential in the antitumor application, but its therapeutic effect is seriously affected by poor water solubility, targeting ability, and low bioavailability. Here, we constructed a novel composite nanomaterial with PCN-224 as a drug carrier and loaded rapamycin, named R@BP@HA. The nanoplate not only improves targeting, but also synergizes rapamycin with PCN-224 to effectively promote tumor cell apoptosis, which subsequently causes immunogenic cell death (ICD), and shows strong therapeutic effect in 4T1 breast cancer model. The treatment effect depends on three main points:(i)Proapoptotic effect of rapamycin on tumor cells;(ii)ROS production by PCN-224-mediated photodynamic therapy;(iii)ICD induced DC maturation, increased immune response and promoted T cell proliferation and differentiation. This nanoplate offers potential antitumor efficacy in combination with chemotherapy, photodynamic therapy, and immunotherapy.
Collapse
Affiliation(s)
- Jihua Tian
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan 030001, China.
| | - Jing Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan 030001, China
| | - Huanyu Xu
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan 030001, China
| | - Bocheng Zou
- Department of The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Weihao Chen
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan 030001, China
| | - Yulong Liu
- Department of The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Jingshu Chen
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan 030001, China
| | - Ruiping Zhang
- Department of The Radiology Department of First Hospital of Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
42
|
Zhang L, Pitcher LE, Prahalad V, Niedernhofer LJ, Robbins PD. Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS J 2023; 290:1362-1383. [PMID: 35015337 DOI: 10.1111/febs.16350] [Citation(s) in RCA: 253] [Impact Index Per Article: 126.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/17/2021] [Accepted: 01/10/2022] [Indexed: 12/15/2022]
Abstract
The concept of geroscience is that since ageing is the greatest risk factor for many diseases and conditions, targeting the ageing process itself will have the greatest impact on human health. Of the hallmarks of ageing, cellular senescence has emerged as a druggable therapeutic target for extending healthspan in model organisms. Cellular senescence is a cell state of irreversible proliferative arrest driven by different types of stress, including oncogene-induced stress. Many senescent cells (SnCs) develop a senescent-associated secretory phenotype (SASP) comprising pro-inflammatory cytokines, chemokines, proteases, bioactive lipids, inhibitory molecules, extracellular vesicles, metabolites, lipids and other factors, able to promote chronic inflammation and tissue dysfunction. SnCs up-regulate senescent cell anti-apoptotic pathways (SCAPs) that prevent them from dying despite the accumulation of damage to DNA and other organelles. These SCAPs and other pathways altered in SnCs represent therapeutic targets for the development of senotherapeutic drugs that induce selective cell death of SnCs, specifically termed senolytics or suppress markers of senescence, in particular the SASP, termed senomorphics. Here, we review the current state of the development of senolytics and senomorphics for the treatment of age-related diseases and disorders and extension of healthy longevity. In addition, the challenges of documenting senolytic and senomorphic activity in pre-clinical models and the current state of the clinical application of the different senotherapeutics will be discussed.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Louise E Pitcher
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Vaishali Prahalad
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Laura J Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Paul D Robbins
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
43
|
Molecular and therapeutic insights of rapamycin: a multi-faceted drug from Streptomyces hygroscopicus. Mol Biol Rep 2023; 50:3815-3833. [PMID: 36696023 PMCID: PMC9875782 DOI: 10.1007/s11033-023-08283-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023]
Abstract
The advancement in pharmaceutical research has led to the discovery and development of new combinatorial life-saving drugs. Rapamycin is a macrolide compound produced from Streptomyces hygroscopicus. Rapamycin and its derivatives are one of the promising sources of drug with broad spectrum applications in the medical field. In recent times, rapamycin has gained significant attention as of its activity against cytokine storm in COVID-19 patients. Rapamycin and its derivatives have more potency when compared to other prevailing drugs. Initially, it has been used exclusively as an anti-fungal drug. Currently rapamycin has been widely used as an immunosuppressant. Rapamycin is a multifaceted drug; it has anti-cancer, anti-viral and anti-aging potentials. Rapamycin has its specific action on mTOR signaling pathway. mTOR has been identified as a key regulator of different pathways. There will be an increased demand for rapamycin, because it has lesser adverse effects when compared to steroids. Currently researchers are focused on the production of effective rapamycin derivatives to combat the growing demand of this wonder drug. The main focus of the current review is to explore the origin, development, molecular mechanistic action, and the current therapeutic aspects of rapamycin. Also, this review article revealed the potential of rapamycin and the progress of rapamycin research. This helps in understanding the exact potency of the drug and could facilitate further studies that could fill in the existing knowledge gaps. The study also gathers significant data pertaining to the gene clusters and biosynthetic pathways involved in the synthesis and production of this multi-faceted drug. In addition, an insight into the mechanism of action of the drug and important derivatives of rapamycin has been expounded. The fillings of the current review, aids in understanding the underlying molecular mechanism, strain improvement, optimization and production of rapamycin derivatives.
Collapse
|
44
|
Yin Y, Zhou Y, Yang X, Xu Z, Yang B, Luo P, Yan H, He Q. The participation of non-canonical autophagic proteins in the autophagy process and their potential as therapeutic targets. Expert Opin Ther Targets 2023; 27:71-86. [PMID: 36735300 DOI: 10.1080/14728222.2023.2177151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Autophagy is a conserved catabolic process that helps recycle intracellular components to maintain homeostasis. The completion of autophagy requires the synergistic effect of multiple canonical autophagic proteins. Defects in autophagy machinery have been reported to promote diseases, rendering autophagy a bone fide health-modifying agent. However, the clinical implication of canonical pan-autophagic activators or inhibitors has often led to undesirable side effects, making it urgent to find a safer autophagy-related therapeutic target. The discovery of non-canonical autophagic proteins has been found to specifically affect the development of diseases without causing a universal impact on autophagy and has shed light on finding a safer way to utilize autophagy in the therapeutic context. AREAS COVERED This review summarizes recently discovered non-canonical autophagic proteins, how these proteins influence autophagy, and their potential therapeutic role in the disease due to their interaction with autophagy. EXPERT OPINION Several therapies have been studied thus far and continued research is needed to identify the potential that non-canonical autophagic proteins have for treating certain diseases. In the meantime, continue to uncover new non-canonical autophagic proteins and examine which are likely to have therapeutic implications.
Collapse
Affiliation(s)
- Yiming Yin
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yourong Zhou
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaochun Yang
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhifei Xu
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Peihua Luo
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Pharmacology and Toxicology, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Yan
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiaojun He
- College of Pharmaceutical Sciences, Center for Drug Safety Evaluation and Research of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
45
|
Baskaran P, Mihaylov SR, Vinsland E, Shah K, Granat L, Ultanir SK, Tee AR, Murn J, Bateman JM. Phosphorylation of the novel mTOR substrate Unkempt regulates cellular morphogenesis. J Biol Chem 2023; 299:102788. [PMID: 36509146 PMCID: PMC9852543 DOI: 10.1016/j.jbc.2022.102788] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a protein kinase that integrates multiple inputs to regulate anabolic cellular processes. For example, mTOR complex 1 (mTORC1) has key functions in growth control, autophagy, and metabolism. However, much less is known about the signaling components that act downstream of mTORC1 to regulate cellular morphogenesis. Here, we show that the RNA-binding protein Unkempt, a key regulator of cellular morphogenesis, is a novel substrate of mTORC1. We show that Unkempt phosphorylation is regulated by nutrient levels and growth factors via mTORC1. To analyze Unkempt phosphorylation, we immunoprecipitated Unkempt from cells in the presence or the absence of the mTORC1 inhibitor rapamycin and used mass spectrometry to identify mTORC1-dependent phosphorylated residues. This analysis showed that mTORC1-dependent phosphorylation is concentrated in a serine-rich intrinsically disordered region in the C-terminal half of Unkempt. We also found that Unkempt physically interacts with and is directly phosphorylated by mTORC1 through binding to the regulatory-associated protein of mTOR, Raptor. Furthermore, analysis in the developing brain of mice lacking TSC1 expression showed that phosphorylation of Unkempt is mTORC1 dependent in vivo. Finally, mutation analysis of key serine/threonine residues in the serine-rich region indicates that phosphorylation inhibits the ability of Unkempt to induce a bipolar morphology. Phosphorylation within this serine-rich region thus profoundly affects the ability of Unkempt to regulate cellular morphogenesis. Taken together, our findings reveal a novel molecular link between mTORC1 signaling and cellular morphogenesis.
Collapse
Affiliation(s)
- Pranetha Baskaran
- Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Simeon R Mihaylov
- Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK; Kinases and Brain Development Lab, The Francis Crick Institute, London, UK
| | - Elin Vinsland
- Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK; Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Kriti Shah
- Department of Biochemistry, University of California, Riverside, California, USA
| | - Lucy Granat
- Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Sila K Ultanir
- Kinases and Brain Development Lab, The Francis Crick Institute, London, UK
| | - Andrew R Tee
- Cancer and Genetics Building, Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park Way, Cardiff, UK
| | - Jernej Murn
- Department of Biochemistry, University of California, Riverside, California, USA.
| | - Joseph M Bateman
- Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK.
| |
Collapse
|
46
|
Liu Z, Zhuang W, Cai M, Lv E, Wang Y, Wu Z, Wang H, Fu W. Kaemperfol Protects Dopaminergic Neurons by Promoting mTOR-Mediated Autophagy in Parkinson’s Disease Models. Neurochem Res 2022; 48:1395-1411. [PMID: 36469163 DOI: 10.1007/s11064-022-03819-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/18/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
We previously showed that kaempferol (KAE) could exert neuroprotective effects against PD. It has been demonstrated that abnormal autophagy plays a key role in the development of PD. Mitochondrial dysfunction, involved in the development of PD, can damage dopaminergic neurons. Whether the protective effects of KAE were exerted via regulating autophagy remains largely undefined, however. This study aimed to investigate whether KAE could protect dopaminergic neurons via autophagy and the underlying mechanisms using a MPTP/MPP+-stimulated PD model. Cell viability was detected by cell counting kit-8 (CCK-8) assay, and protein levels of autophagy mediators along with mTOR signaling pathway molecules were investigated by immunohistochemistry and Western blot analyses. The results showed that KAE could ameliorate the behavioral impairments of mice, reduce the loss of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra pars compacta, and reduce α-synuclein (α-syn) levels. Furthermore, KAE upregulated levels of autophagy effector protein of Beclin-1 and autophagy microtubule associated protein of light chain 3 (LC3) in the substantia nigra (SN) while rescuing mitochondrial integrity, and downregulated levels of ubiquitin binding protein p62 and cleaved caspase-3, probably by decreasing the mammalian target of rapamycin (mTOR) signaling pathway. Further in vitro experiments demonstrated similar results. In conclusion, KAE exerts neuroprotective effects against PD potentially by promoting autophagy via inhibiting the mTOR signaling pathway.
Collapse
Affiliation(s)
- Zhan Liu
- Department of Histology and Embryology, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Wenxin Zhuang
- Center for Experimental Medical Research, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Meiyun Cai
- Department of Histology and Embryology, Weifang Medical University, Weifang, 261053, Shandong, China
| | - E Lv
- Department of Histology and Embryology, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Yanqiang Wang
- Department of Neurology, Affiliated Hospital of Weifang Medical University, Weifang, 261053, Shandong, China
| | - Zhengyan Wu
- Department of Biotechnology, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Hongyu Wang
- Department of Biotechnology, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Wenyu Fu
- Department of Histology and Embryology, Weifang Medical University, Weifang, 261053, Shandong, China.
| |
Collapse
|
47
|
Sherif S, Roelands J, Mifsud W, Ahmed EI, Raynaud CM, Rinchai D, Sathappan A, Maaz A, Saleh A, Ozer E, Fakhro KA, Mifsud B, Thorsson V, Bedognetti D, Hendrickx WRL. The immune landscape of solid pediatric tumors. J Exp Clin Cancer Res 2022; 41:199. [PMID: 35690832 PMCID: PMC9188257 DOI: 10.1186/s13046-022-02397-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/18/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Large immunogenomic analyses have demonstrated the prognostic role of the functional orientation of the tumor microenvironment in adult solid tumors, this variable has been poorly explored in the pediatric counterpart.
Methods
We performed a systematic analysis of public RNAseq data (TARGET) for five pediatric tumor types (408 patients): Wilms tumor (WLM), neuroblastoma (NBL), osteosarcoma (OS), clear cell sarcoma of the kidney (CCSK) and rhabdoid tumor of the kidney (RT). We assessed the performance of the Immunologic Constant of Rejection (ICR), which captures an active Th1/cytotoxic response. We also performed gene set enrichment analysis (ssGSEA) and clustered more than 100 well characterized immune traits to define immune subtypes and compared their outcome.
Results
A higher ICR score was associated with better survival in OS and high risk NBL without MYCN amplification but with poorer survival in WLM. Clustering of immune traits revealed the same five principal modules previously described in adult tumors (TCGA). These modules divided pediatric patients into six immune subtypes (S1-S6) with distinct survival outcomes. The S2 cluster showed the best overall survival, characterized by low enrichment of the wound healing signature, high Th1, and low Th2 infiltration, while the reverse was observed in S4. Upregulation of the WNT/Beta-catenin pathway was associated with unfavorable outcomes and decreased T-cell infiltration in OS.
Conclusions
We demonstrated that extracranial pediatric tumors could be classified according to their immune disposition, unveiling similarities with adults’ tumors. Immunological parameters might be explored to refine diagnostic and prognostic biomarkers and to identify potential immune-responsive tumors.
Collapse
|
48
|
Kaiser MS, Milan G, Ham DJ, Lin S, Oliveri F, Chojnowska K, Tintignac LA, Mittal N, Zimmerli CE, Glass DJ, Zavolan M, Rüegg MA. Dual roles of mTORC1-dependent activation of the ubiquitin-proteasome system in muscle proteostasis. Commun Biol 2022; 5:1141. [PMID: 36302954 PMCID: PMC9613904 DOI: 10.1038/s42003-022-04097-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/11/2022] [Indexed: 12/02/2022] Open
Abstract
Muscle size is controlled by the PI3K-PKB/Akt-mTORC1-FoxO pathway, which integrates signals from growth factors, energy and amino acids to activate protein synthesis and inhibit protein breakdown. While mTORC1 activity is necessary for PKB/Akt-induced muscle hypertrophy, its constant activation alone induces muscle atrophy. Here we show that this paradox is based on mTORC1 activity promoting protein breakdown through the ubiquitin-proteasome system (UPS) by simultaneously inducing ubiquitin E3 ligase expression via feedback inhibition of PKB/Akt and proteasome biogenesis via Nuclear Factor Erythroid 2-Like 1 (Nrf1). Muscle growth was restored by reactivation of PKB/Akt, but not by Nrf1 knockdown, implicating ubiquitination as the limiting step. However, both PKB/Akt activation and proteasome depletion by Nrf1 knockdown led to an immediate disruption of proteome integrity with rapid accumulation of damaged material. These data highlight the physiological importance of mTORC1-mediated PKB/Akt inhibition and point to juxtaposed roles of the UPS in atrophy and proteome integrity. Exploring the relationship between mTORC1 and the ubiquitin-proteasome system, light is shed on the paradox between mTORC1-mediated muscle hypertrophy induced by PKB/Akt and the muscle atrophy induced by mTORC1 alone.
Collapse
Affiliation(s)
- Marco S Kaiser
- Biozentrum, University of Basel, Basel, Switzerland.,BIOREBA AG, Christoph Merian-Ring 7, 4153, Reinach, Switzerland
| | - Giulia Milan
- Biozentrum, University of Basel, Basel, Switzerland. .,Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland.
| | - Daniel J Ham
- Biozentrum, University of Basel, Basel, Switzerland.
| | - Shuo Lin
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - Kathrin Chojnowska
- Biozentrum, University of Basel, Basel, Switzerland.,AstraZeneca AG, Neuhofstrasse 34, 6340, Baar, Switzerland
| | - Lionel A Tintignac
- Biozentrum, University of Basel, Basel, Switzerland.,Neuromuscular Research Group, Departments of Neurology and Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | | | - Christian E Zimmerli
- Biozentrum, University of Basel, Basel, Switzerland.,Department of Molecular Sociology, Max Planck Institute of Biophysics, Max-von-Laue-Straße 3, 60438, Frankfurt am Main, Germany
| | - David J Glass
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA.,Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | |
Collapse
|
49
|
Franz T, Negele J, Bruno P, Böttcher M, Mitchell-Flack M, Reemts L, Krone A, Mougiakakos D, Müller AJ, Zautner AE, Kahlfuss S. Pleiotropic effects of antibiotics on T cell metabolism and T cell-mediated immunity. Front Microbiol 2022; 13:975436. [DOI: 10.3389/fmicb.2022.975436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
T cells orchestrate adaptive and innate immune responses against pathogens and transformed cells. However, T cells are also the main adaptive effector cells that mediate allergic and autoimmune reactions. Within the last few years, it has become abundantly clear that activation, differentiation, effector function, and environmental adaptation of T cells is closely linked to their energy metabolism. Beyond the provision of energy equivalents, metabolic pathways in T cells generate building blocks required for clonal expansion. Furthermore, metabolic intermediates directly serve as a source for epigenetic gene regulation by histone and DNA modification mechanisms. To date, several antibiotics were demonstrated to modulate the metabolism of T cells especially by altering mitochondrial function. Here, we set out to systematically review current evidence about how beta-lactam antibiotics, macrolides, fluoroquinolones, tetracyclines, oxazolidinones, nitroimidazoles, and amphenicols alter the metabolism and effector functions of CD4+ T helper cell populations and CD8+ T cells in vitro and in vivo. Based on this evidence, we have developed an overview on how the use of these antibiotics may be beneficial or detrimental in T cell-mediated physiological and pathogenic immune responses, such as allergic and autoimmune diseases, by altering the metabolism of different T cell populations.
Collapse
|
50
|
Recent Advances in the Aging Microenvironment of Breast Cancer. Cancers (Basel) 2022; 14:cancers14204990. [PMID: 36291773 PMCID: PMC9599409 DOI: 10.3390/cancers14204990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary The incidence of breast cancer has increased rapidly in recent years. Aging is one of the risk factors for advanced breast cancer. More and more studies have been conducted on the influence of the aging microenvironment on breast cancer. In this review, we summarize the effects of physical changes in the aging microenvironment, senescence-associated secretory phenotypes, and senescent stromal cells on the initiation and progression of breast cancer and the underlying mechanisms. In addition, we also discuss potential targets for senotherapeutics and senescence-inducing agents in the aging microenvironment of breast cancer. We hope this review can provide some directions for future research on the aging microenvironment in breast cancer. Abstract Aging is one of the risk factors for advanced breast cancer. With the increasing trend toward population aging, it is important to study the effects of aging on breast cancer in depth. Cellular senescence and changes in the aging microenvironment in vivo are the basis for body aging and death. In this review, we focus on the influence of the aging microenvironment on breast cancer. Increased breast extracellular matrix stiffness in the aging breast extracellular matrix can promote the invasion of breast cancer cells. The role of senescence-associated secretory phenotypes (SASPs) such as interleukin-6 (IL-6), IL-8, and matrix metalloproteases (MMPs), in breast cancer cell proliferation, invasion, and metastasis is worthy of exploration. Furthermore, the impact of senescent fibroblasts, adipocytes, and endothelial cells on the mammary matrix is discussed in detail. We also list potential targets for senotherapeutics and senescence-inducing agents in the aging microenvironment of breast cancer. In conclusion, this review offers an overview of the influence of the aging microenvironment on breast cancer initiation and progression, with the aim of providing some directions for future research on the aging microenvironment in breast cancer.
Collapse
|