1
|
Duse DA, Schröder NH, Srivastava T, Benkhoff M, Vogt J, Nowak MK, Funk F, Semleit N, Wollnitzke P, Erkens R, Kötter S, Meuth SG, Keul P, Santos W, Polzin A, Kelm M, Krüger M, Schmitt J, Levkau B. Deficiency of the sphingosine-1-phosphate (S1P) transporter Mfsd2b protects the heart against hypertension-induced cardiac remodeling by suppressing the L-type-Ca 2+ channel. Basic Res Cardiol 2024; 119:853-868. [PMID: 39110173 PMCID: PMC11461684 DOI: 10.1007/s00395-024-01073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 10/09/2024]
Abstract
The erythrocyte S1P transporter Mfsd2b is also expressed in the heart. We hypothesized that S1P transport by Mfsd2b is involved in cardiac function. Hypertension-induced cardiac remodeling was induced by 4-weeks Angiotensin II (AngII) administration and assessed by echocardiography. Ca2+ transients and sarcomere shortening were examined in adult cardiomyocytes (ACM) from Mfsd2b+/+ and Mfsd2b-/- mice. Tension and force development were measured in skinned cardiac fibers. Myocardial gene expression was determined by real-time PCR, Protein Phosphatase 2A (PP2A) by enzymatic assay, and S1P by LC/MS, respectively. Msfd2b was expressed in the murine and human heart, and its deficiency led to higher cardiac S1P. Mfsd2b-/- mice had regular basal cardiac function but were protected against AngII-induced deterioration of left-ventricular function as evidenced by ~ 30% better stroke volume and cardiac index, and preserved ejection fraction despite similar increases in blood pressure. Mfsd2b-/- ACM exhibited attenuated Ca2+ mobilization in response to isoprenaline whereas contractility was unchanged. Mfsd2b-/- ACM showed no changes in proteins responsible for Ca2+ homeostasis, and skinned cardiac fibers exhibited reduced passive tension generation with preserved contractility. Verapamil abolished the differences in Ca2+ mobilization between Mfsd2b+/+ and Mfsd2b-/- ACM suggesting that S1P inhibits L-type-Ca2+ channels (LTCC). In agreement, intracellular S1P activated the inhibitory LTCC phosphatase PP2A in ACM and PP2A activity was increased in Mfsd2b-/- hearts. We suggest that myocardial S1P protects from hypertension-induced left-ventricular remodeling by inhibiting LTCC through PP2A activation. Pharmacologic inhibition of Mfsd2b may thus offer a novel approach to heart failure.
Collapse
Affiliation(s)
- Dragos Andrei Duse
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Nathalie Hannelore Schröder
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Tanu Srivastava
- Institute of Pharmacology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Marcel Benkhoff
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Jens Vogt
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Melissa Kim Nowak
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Florian Funk
- Institute of Pharmacology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Nina Semleit
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Philipp Wollnitzke
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Ralf Erkens
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Sebastian Kötter
- Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sven Günther Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - Petra Keul
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany
| | - Webster Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA, 24060, USA
| | - Amin Polzin
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pneumology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Martina Krüger
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
- Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Joachim Schmitt
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
- Institute of Pharmacology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Bodo Levkau
- Institute for Molecular Medicine III, University Hospital Düsseldorf and Heinrich Heine University, Düsseldorf, Germany.
- Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany.
| |
Collapse
|
2
|
Choi JE, Qiao Y, Kryczek I, Yu J, Gurkan J, Bao Y, Gondal M, Tien JCY, Maj T, Yazdani S, Parolia A, Xia H, Zhou J, Wei S, Grove S, Vatan L, Lin H, Li G, Zheng Y, Zhang Y, Cao X, Su F, Wang R, He T, Cieslik M, Green MD, Zou W, Chinnaiyan AM. PIKfyve controls dendritic cell function and tumor immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.28.582543. [PMID: 38464258 PMCID: PMC10925294 DOI: 10.1101/2024.02.28.582543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The modern armamentarium for cancer treatment includes immunotherapy and targeted therapy, such as protein kinase inhibitors. However, the mechanisms that allow cancer-targeting drugs to effectively mobilize dendritic cells (DCs) and affect immunotherapy are poorly understood. Here, we report that among shared gene targets of clinically relevant protein kinase inhibitors, high PIKFYVE expression was least predictive of complete response in patients who received immune checkpoint blockade (ICB). In immune cells, high PIKFYVE expression in DCs was associated with worse response to ICB. Genetic and pharmacological studies demonstrated that PIKfyve ablation enhanced DC function via selectively altering the alternate/non-canonical NF-κB pathway. Both loss of Pikfyve in DCs and treatment with apilimod, a potent and specific PIKfyve inhibitor, restrained tumor growth, enhanced DC-dependent T cell immunity, and potentiated ICB efficacy in tumor-bearing mouse models. Furthermore, the combination of a vaccine adjuvant and apilimod reduced tumor progression in vivo. Thus, PIKfyve negatively controls DCs, and PIKfyve inhibition has promise for cancer immunotherapy and vaccine treatment strategies.
Collapse
Affiliation(s)
- Jae Eun Choi
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuanyuan Qiao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Jiali Yu
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Gurkan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yi Bao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mahnoor Gondal
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jean Ching-Yi Tien
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tomasz Maj
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Sahr Yazdani
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Houjun Xia
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - JiaJia Zhou
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Heng Lin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Gaopeng Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Yang Zheng
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuping Zhang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xuhong Cao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fengyun Su
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rui Wang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tongchen He
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Marcin Cieslik
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Michael D. Green
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Radiation Oncology Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Arul M. Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
3
|
Choi JE, Qiao Y, Kryczek I, Yu J, Gurkan J, Bao Y, Gondal M, Tien JCY, Maj T, Yazdani S, Parolia A, Xia H, Zhou J, Wei S, Grove S, Vatan L, Lin H, Li G, Zheng Y, Zhang Y, Cao X, Su F, Wang R, He T, Cieslik M, Green MD, Zou W, Chinnaiyan AM. PIKfyve, expressed by CD11c-positive cells, controls tumor immunity. Nat Commun 2024; 15:5487. [PMID: 38942798 PMCID: PMC11213953 DOI: 10.1038/s41467-024-48931-9] [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: 08/17/2023] [Accepted: 05/15/2024] [Indexed: 06/30/2024] Open
Abstract
Cancer treatment continues to shift from utilizing traditional therapies to targeted ones, such as protein kinase inhibitors and immunotherapy. Mobilizing dendritic cells (DC) and other myeloid cells with antigen presenting and cancer cell killing capacities is an attractive but not fully exploited approach. Here, we show that PIKFYVE is a shared gene target of clinically relevant protein kinase inhibitors and high expression of this gene in DCs is associated with poor patient response to immune checkpoint blockade (ICB) therapy. Genetic and pharmacological studies demonstrate that PIKfyve ablation enhances the function of CD11c+ cells (predominantly dendritic cells) via selectively altering the non-canonical NF-κB pathway. Both loss of Pikfyve in CD11c+ cells and treatment with apilimod, a potent and specific PIKfyve inhibitor, restrained tumor growth, enhanced DC-dependent T cell immunity, and potentiated ICB efficacy in tumor-bearing mouse models. Furthermore, the combination of a vaccine adjuvant and apilimod reduced tumor progression in vivo. Thus, PIKfyve negatively regulates the function of CD11c+ cells, and PIKfyve inhibition has promise for cancer immunotherapy and vaccine treatment strategies.
Collapse
Affiliation(s)
- Jae Eun Choi
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Yuanyuan Qiao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Jiali Yu
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Gurkan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yi Bao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mahnoor Gondal
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jean Ching-Yi Tien
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tomasz Maj
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Sahr Yazdani
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Houjun Xia
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - JiaJia Zhou
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Heng Lin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Gaopeng Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
| | - Yang Zheng
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yuping Zhang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Xuhong Cao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fengyun Su
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rui Wang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Tongchen He
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Marcin Cieslik
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Michael D Green
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Department of Radiation Oncology Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Weiping Zou
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, MI, USA.
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Urology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Lu Z, Lai Q, Li ZF, Zhong MY, Jiang YL, Feng LY, Zha J, Yao JW, Li Y, Deng XM, Xu B. Novel PIKfyve/Tubulin Dual-target Inhibitor as a Promising Therapeutic Strategy for B-cell Acute Lymphoblastic Leukemia. Curr Med Sci 2024; 44:298-308. [PMID: 38619682 DOI: 10.1007/s11596-024-2847-5] [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: 05/24/2023] [Accepted: 01/19/2024] [Indexed: 04/16/2024]
Abstract
OBJECTIVE In B-cell acute lymphoblastic leukemia (B-ALL), current intensive chemotherapies for adult patients fail to achieve durable responses in more than 50% of cases, underscoring the urgent need for new therapeutic regimens for this patient population. The present study aimed to determine whether HZX-02-059, a novel dual-target inhibitor targeting both phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) and tubulin, is lethal to B-ALL cells and is a potential therapeutic for B-ALL patients. METHODS Cell proliferation, vacuolization, apoptosis, cell cycle, and in-vivo tumor growth were evaluated. In addition, Genome-wide RNA-sequencing studies were conducted to elucidate the mechanisms of action underlying the anti-leukemia activity of HZX-02-059 in B-ALL. RESULTS HZX-02-059 was found to inhibit cell proliferation, induce vacuolization, promote apoptosis, block the cell cycle, and reduce in-vivo tumor growth. Downregulation of the p53 pathway and suppression of the phosphoinositide 3-kinase (PI3K)/AKT pathway and the downstream transcription factors c-Myc and NF-κB were responsible for these observations. CONCLUSION Overall, these findings suggest that HZX-02-059 is a promising agent for the treatment of B-ALL patients resistant to conventional therapies.
Collapse
Affiliation(s)
- Zhen Lu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361005, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361005, China
| | - Qian Lai
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361005, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361005, China
| | - Zhi-Feng Li
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361005, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361005, China
| | - Meng-Ya Zhong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361005, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361005, China
| | - Yue-Long Jiang
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361005, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361005, China
| | - Li-Ying Feng
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361005, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361005, China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361005, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361005, China
| | - Jing-Wei Yao
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361005, China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361005, China
| | - Yin Li
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510630, China.
| | - Xian-Ming Deng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361003, China.
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361005, China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361005, China.
| |
Collapse
|
5
|
Wible DJ, Parikh Z, Cho EJ, Chen MD, Jeter CR, Mukhopadhyay S, Dalby KN, Varadarajan S, Bratton SB. Unexpected inhibition of the lipid kinase PIKfyve reveals an epistatic role for p38 MAPKs in endolysosomal fission and volume control. Cell Death Dis 2024; 15:80. [PMID: 38253602 PMCID: PMC10803372 DOI: 10.1038/s41419-024-06423-0] [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: 05/09/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024]
Abstract
p38 mitogen-activated protein kinases (MAPKs) participate in autophagic signaling; and previous reports suggest that pyridinyl imidazole p38 MAPK inhibitors, including SB203580 and SB202190, induce cell death in some cancer cell-types through unrestrained autophagy. Subsequent studies, however, have suggested that the associated cytoplasmic vacuolation resulted from off-target inhibition of an unidentified enzyme. Herein, we report that SB203580-induced vacuolation is rapid, reversible, and relies on the class III phosphatidylinositol 3-kinase (PIK3C3) complex and the production of phosphatidylinositol 3-phosphate [PI(3)P] but not on autophagy per se. Rather, vacuolation resulted from the accumulation of Rab7 on late endosome and lysosome (LEL) membranes, combined with an osmotic imbalance that triggered severe swelling in these organelles. Inhibition of PIKfyve, the lipid kinase that converts PI(3)P to PI(3,5)P2 on LEL membranes, produced a similar phenotype in cells; therefore, we performed in vitro kinase assays and discovered that both SB203580 and SB202190 directly inhibited recombinant PIKfyve. Cancer cells treated with either drug likewise displayed significant reductions in the endogenous levels of PI(3,5)P2. Despite these results, SB203580-induced vacuolation was not entirely due to off-target inhibition of PIKfyve, as a drug-resistant p38α mutant suppressed vacuolation; and combined genetic deletion of both p38α and p38β dramatically sensitized cells to established PIKfyve inhibitors, including YM201636 and apilimod. The rate of vacuole dissolution (i.e., LEL fission), following the removal of apilimod, was also significantly reduced in cells treated with BIRB-796, a structurally unrelated p38 MAPK inhibitor. Thus, our studies indicate that pyridinyl imidazole p38 MAPK inhibitors induce cytoplasmic vacuolation through the combined inhibition of both PIKfyve and p38 MAPKs, and more generally, that p38 MAPKs act epistatically to PIKfyve, most likely to promote LEL fission.
Collapse
Affiliation(s)
- Daric J Wible
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Zalak Parikh
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Eun Jeong Cho
- Targeted Therapeutic Drug Discovery and Development Program, Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Miao-Der Chen
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Collene R Jeter
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Somshuvra Mukhopadhyay
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Kevin N Dalby
- Targeted Therapeutic Drug Discovery and Development Program, Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Shankar Varadarajan
- Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Shawn B Bratton
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.
| |
Collapse
|
6
|
Thomas N, Schröder NH, Nowak MK, Wollnitzke P, Ghaderi S, von Wnuck Lipinski K, Wille A, Deister-Jonas J, Vogt J, Gräler MH, Dannenberg L, Buschmann T, Westhoff P, Polzin A, Kelm M, Keul P, Weske S, Levkau B. Sphingosine-1-phosphate suppresses GLUT activity through PP2A and counteracts hyperglycemia in diabetic red blood cells. Nat Commun 2023; 14:8329. [PMID: 38097610 PMCID: PMC10721873 DOI: 10.1038/s41467-023-44109-x] [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: 11/30/2023] [Indexed: 12/17/2023] Open
Abstract
Red blood cells (RBC) are the major carriers of sphingosine-1-phosphate (S1P) in blood. Here we show that variations in RBC S1P content achieved by altering S1P synthesis and transport by genetic and pharmacological means regulate glucose uptake and metabolic flux. This is due to S1P-mediated activation of the catalytic protein phosphatase 2 (PP2A) subunit leading to reduction of cell-surface glucose transporters (GLUTs). The mechanism dynamically responds to metabolic cues from the environment by increasing S1P synthesis, enhancing PP2A activity, reducing GLUT phosphorylation and localization, and diminishing glucose uptake in RBC from diabetic mice and humans. Functionally, it protects RBC against lipid peroxidation in hyperglycemia and diabetes by activating the pentose phosphate pathway. Proof of concept is provided by the resistance of mice lacking the S1P exporter MFSD2B to diabetes-induced HbA1c elevation and thiobarbituric acid reactive substances (TBARS) generation in diabetic RBC. This mechanism responds to pharmacological S1P analogues such as fingolimod and may be functional in other insulin-independent tissues making it a promising therapeutic target.
Collapse
Affiliation(s)
- Nadine Thomas
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | - Nathalie H Schröder
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | - Melissa K Nowak
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | - Philipp Wollnitzke
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | - Shahrooz Ghaderi
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | | | - Annalena Wille
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | | | - Jens Vogt
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
- Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Lisa Dannenberg
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Tobias Buschmann
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | - Philipp Westhoff
- Institute of Plant Biochemistry, Cluster of Excellence on Plant Sciences (CEPLAS), Heinrich Heine University, Düsseldorf, Germany
| | - Amin Polzin
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Petra Keul
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | - Sarah Weske
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, Heinrich Heine University, Düsseldorf, Germany.
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany.
| |
Collapse
|
7
|
Hwang N, Yoon BK, Chun KH, Kim H, Lee Y, Kim JW, Jeon H, Kim TH, Kim MY, Fang S, Cheong JH, Kim JW. Caveolin-1 mediates the utilization of extracellular proteins for survival in refractory gastric cancer. Exp Mol Med 2023; 55:2461-2472. [PMID: 37919422 PMCID: PMC10689497 DOI: 10.1038/s12276-023-01109-7] [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: 04/30/2023] [Revised: 07/20/2023] [Accepted: 07/31/2023] [Indexed: 11/04/2023] Open
Abstract
Despite advances in cancer therapy, the clinical outcome of patients with gastric cancer remains poor, largely due to tumor heterogeneity. Thus, finding a hidden vulnerability of clinically refractory subtypes of gastric cancer is crucial. Here, we report that chemoresistant gastric cancer cells rely heavily on endocytosis, facilitated by caveolin-1, for survival. caveolin-1 was highly upregulated in the most malignant stem-like/EMT/mesenchymal (SEM)-type gastric cancer cells, allowing caveolin-1-mediated endocytosis and utilization of extracellular proteins via lysosomal degradation. Downregulation of caveolin-1 alone was sufficient to induce cell death in SEM-type gastric cancer cells, emphasizing its importance as a survival mechanism. Consistently, chloroquine, a lysosomal inhibitor, successfully blocked caveolin-1-mediated endocytosis, leading to the marked suppression of tumor growth in chemorefractory gastric cancer cells in vitro, including patient-derived organoids, and in vivo. Together, our findings suggest that caveolin-1-mediated endocytosis is a key metabolic pathway for gastric cancer survival and a potential therapeutic target.
Collapse
Affiliation(s)
- Nahee Hwang
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bo Kyung Yoon
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyu-Hye Chun
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyeonhui Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoseob Lee
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Won Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyeonuk Jeon
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Hyun Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mi-Young Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Severance Biomedical Science Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Jae-Ho Cheong
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Department of R&D, Veraverse Inc., Seoul, Republic of Korea.
| | - Jae-Woo Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Liu X, Ma M, Tian S, Wang W, Li X. "Domino" cascade reactor based on DNA hydrogel for synergistic treatment of malignant tumor. Eur J Med Chem 2023; 256:115441. [PMID: 37182333 DOI: 10.1016/j.ejmech.2023.115441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 05/16/2023]
Abstract
The action pathways of starvation therapy and photodynamic therapy (PDT) do not exist in isolation and are usually related to tumor cell metabolism and immune regulation, which are of great significance in the treatment of malignant tumors. Here, a cancer-targeted "domino" cascade reactor is constructed for synergistic starvation therapy and amplifies photodynamic therapy by assembling hemin and glucose oxidase (GOx) into DNA hydrogel load with hypoxia-inducible factor 1α (HIF-1α) and photosensitizer chlorin e6 (Ce6). The cascade reactor has excellent biocompatibility and tumor targeting, which promotes PDT by reducing HIF-1α. At the same time, the G-quadruplex of AS1411 combined with hemin (AH) catalyzes the generation of oxygen from hydrogen peroxide to further improve the efficiency of PDT. The synergistic therapeutic effect of the cascade reactor is evaluated through in vivo and in vitro experiments, indicating that this cascade reactor has great potential advantages in the synergistic treatment of cancer.
Collapse
Affiliation(s)
- Xiaofan Liu
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, China
| | - Minghui Ma
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, China
| | - Shuo Tian
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, China
| | - Weicai Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, China
| | - Xuemei Li
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumor Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, China.
| |
Collapse
|
9
|
Wible DJ, Parikh Z, Cho EJ, Chen MD, Mukhopadhyay S, Dalby KN, Varadarajan S, Bratton SB. Unexpected inhibition of the lipid kinase PIKfyve reveals an epistatic role for p38 MAPKs in endolysosomal fission and volume control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532495. [PMID: 36993747 PMCID: PMC10054966 DOI: 10.1101/2023.03.13.532495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
p38 mitogen-activated protein kinases (MAPKs) regulate early endocytic trafficking, but their effects on late endocytic trafficking remain unclear. Herein, we report that the pyridinyl imidazole p38 MAPK inhibitors, SB203580 and SB202190, induce a rapid but reversible Rab7-dependent accumulation of large cytoplasmic vacuoles. While SB203580 did not induce canonical autophagy, phosphatidylinositol 3-phosphate [PI(3)P] accumulated on vacuole membranes, and inhibition of the class III PI3-kinase (PIK3C3/VPS34) suppressed vacuolation. Ultimately, vacuolation resulted from the fusion of ER/Golgi-derived membrane vesicles with late endosomes and lysosomes (LELs), combined with an osmotic imbalance in LELs that led to severe swelling and a decrease in LEL fission. Since PIKfyve inhibitors induce a similar phenotype by preventing the conversion of PI(3)P to PI(3,5)P2, we performed in vitro kinase assays and found that PIKfyve activity was unexpectedly inhibited by SB203580 and SB202190, corresponding to losses in endogenous PI(3,5)P2 levels in treated cells. However, vacuolation was not entirely due to 'off-target' inhibition of PIKfyve by SB203580, as a drug-resistant p38α mutant suppressed vacuolation. Moreover, genetic deletion of both p38α and p38β rendered cells dramatically more sensitive to PIKfyve inhibitors, including YM201636 and apilimod. In subsequent 'washout' experiments, the rate of vacuole dissolution upon the removal of apilimod was also significantly reduced in cells treated with BIRB-796, a structurally unrelated p38 MAPK inhibitor. Thus, p38 MAPKs act epistatically to PIKfyve to promote LEL fission; and pyridinyl imidazole p38 MAPK inhibitors induce cytoplasmic vacuolation through the combined inhibition of both PIKfyve and p38 MAPKs.
Collapse
|
10
|
Finicle B, Eckenstein K, Revenko A, Anderson B, Wan W, McCracken A, Gil D, Fruman D, Hanessian S, Seth P, Edinger A. Simultaneous inhibition of endocytic recycling and lysosomal fusion sensitizes cells and tissues to oligonucleotide therapeutics. Nucleic Acids Res 2023; 51:1583-1599. [PMID: 36727438 PMCID: PMC9976930 DOI: 10.1093/nar/gkad023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 02/03/2023] Open
Abstract
Inefficient endosomal escape remains the primary barrier to the broad application of oligonucleotide therapeutics. Liver uptake after systemic administration is sufficiently robust that a therapeutic effect can be achieved but targeting extrahepatic tissues remains challenging. Prior attempts to improve oligonucleotide activity using small molecules that increase the leakiness of endosomes have failed due to unacceptable toxicity. Here, we show that the well-tolerated and orally bioavailable synthetic sphingolipid analog, SH-BC-893, increases the activity of antisense oligonucleotides (ASOs) and small interfering RNAs (siRNAs) up to 200-fold in vitro without permeabilizing endosomes. SH-BC-893 treatment trapped endocytosed oligonucleotides within extra-lysosomal compartments thought to be more permeable due to frequent membrane fission and fusion events. Simultaneous disruption of ARF6-dependent endocytic recycling and PIKfyve-dependent lysosomal fusion was necessary and sufficient for SH-BC-893 to increase non-lysosomal oligonucleotide levels and enhance their activity. In mice, oral administration of SH-BC-893 increased ASO potency in the liver by 15-fold without toxicity. More importantly, SH-BC-893 enabled target RNA knockdown in the CNS and lungs of mice treated subcutaneously with cholesterol-functionalized duplexed oligonucleotides or unmodified ASOs, respectively. Together, these results establish the feasibility of using a small molecule that disrupts endolysosomal trafficking to improve the activity of oligonucleotides in extrahepatic tissues.
Collapse
Affiliation(s)
- Brendan T Finicle
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Kazumi H Eckenstein
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | | | | | - W Brad Wan
- Ionis Pharmaceuticals, Carlsbad, CA, USA
| | | | | | - David A Fruman
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
| | - Stephen Hanessian
- Department of Chemistry, Université de Montréal, Montréal, QC, Canada
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| | | | - Aimee L Edinger
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
11
|
Ghezzi C, Chen BY, Damoiseaux R, Clark PM. Pacritinib inhibits glucose consumption in squamous cell lung cancer cells by targeting FLT3. Sci Rep 2023; 13:1442. [PMID: 36697489 PMCID: PMC9876922 DOI: 10.1038/s41598-023-28576-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Squamous cell lung cancer maintains its growth through elevated glucose consumption, but selective glucose consumption inhibitors are lacking. Here, we discovered using a high-throughput screen new compounds that block glucose consumption in three squamous cell lung cancer cell lines and identified 79 compounds that block glucose consumption in one or more of these cell lines. Based on its ability to block glucose consumption in all three cell lines, pacritinib, an inhibitor of FMS Related Receptor Tyrosine Kinase 3 (FLT3) and Janus Kinase 2 (JAK2), was further studied. Pacritinib decreased glucose consumption in squamous cell lung cancer cells in cell culture and in vivo without affecting glucose consumption in healthy tissues. Pacritinib blocked hexokinase activity, and Hexokinase 1 and 2 mRNA and protein expression. Overexpression of Hexokinase 1 blocked the ability of pacritinib to inhibit glucose consumption in squamous cell lung cancer cells. Overexpression of FLT3 but not JAK2 significantly increased glucose consumption and blocked the ability of pacritinib to inhibit glucose consumption in squamous cell lung cancer cells. Additional FLT3 inhibitors blocked glucose consumption in squamous cell lung cancer cells. Our study identifies FLT3 inhibitors as a new class of inhibitors that can block glucose consumption in squamous cell lung cancer.
Collapse
Affiliation(s)
- Chiara Ghezzi
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Bao Ying Chen
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Robert Damoiseaux
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Peter M Clark
- Crump Institute for Molecular Imaging, University of California, Los Angeles, Box 951770, Los Angeles, CA, 90095, USA.
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
12
|
Wang W, Wang Y, Ma M, Jin HJ, Li X. Drug-Induced Self-Assembly Cascade Nanoreactor for Synergistic Tumor Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:44029-44038. [PMID: 36153981 DOI: 10.1021/acsami.2c09947] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The construction of completely biocompatible and biodegradable tumor suppressors by a simple and reliable method is essential for the clinical application of cancer-targeted drugs. Herein, by inserting glucose oxidase (GOx), catalase (CAT), and chlorin e6 (Ce6) into human serum albumin (HSA) assembly molecules, we constructed a cancer-targeted cascade bioreactor for synergistic starvation and photodynamic therapy (PDT). The modification of HSA could block the GOx activity and reduce the cytotoxicity of normal cells and organs. Through active targeting and passive enhanced permeability and retention effect, the loading of AS1411 could promote the cascade bioreactors to effectively target nucleolin-overexpressed tumors. Once internalized by cancer cells, as a result of catalyzing hydrogen peroxide (H2O2) to produce oxygen (O2), the protein nano-cascade reactor promoted microenvironmental oxygenation, which would subsequently lead to an increase in cytotoxic singlet oxygen (1O2) production under light irradiation as well as the decomposition of intracellular glucose. In vitro and in vivo studies showed that the cascaded nanoreactors could significantly enhance therapeutic efficacy through synergistic starvation therapy and enhanced PDT as well as chemotherapy. This cascade strategy will be demonstrated in clinical applications with huge potential.
Collapse
Affiliation(s)
- Weicai Wang
- Collaborative Innovation Centre of Tumour Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
- Department of Bioscience and Biotechnology, The University of Suwon, Hwaseong City, Gyeonggi-Do 18323, Republic of Korea
| | - Yuanyuan Wang
- Collaborative Innovation Centre of Tumour Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Minghui Ma
- Collaborative Innovation Centre of Tumour Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Hyung Jong Jin
- Department of Bioscience and Biotechnology, The University of Suwon, Hwaseong City, Gyeonggi-Do 18323, Republic of Korea
| | - Xuemei Li
- Collaborative Innovation Centre of Tumour Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Province Key Laboratory of Detection Technology for Tumour Makers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| |
Collapse
|
13
|
Yu W, Wang Z, Yu X, Zhao Y, Xie Z, Zhang K, Chi Z, Chen S, Xu T, Jiang D, Guo X, Li M, Zhang J, Fang H, Yang D, Guo Y, Yang X, Zhang X, Wu Y, Yang W, Wang D. Kir2.1-mediated membrane potential promotes nutrient acquisition and inflammation through regulation of nutrient transporters. Nat Commun 2022; 13:3544. [PMID: 35729093 PMCID: PMC9213538 DOI: 10.1038/s41467-022-31149-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 05/26/2022] [Indexed: 12/22/2022] Open
Abstract
Immunometabolism contributes to inflammation, but how activated macrophages acquire extracellular nutrients to fuel inflammation is largely unknown. Here, we show that the plasma membrane potential (Vm) of macrophages mediated by Kir2.1, an inwardly-rectifying K+ channel, is an important determinant of nutrient acquisition and subsequent metabolic reprogramming promoting inflammation. In the absence of Kir2.1 activity, depolarized macrophage Vm lead to a caloric restriction state by limiting nutrient uptake and concomitant adaptations in nutrient conservation inducing autophagy, AMPK (Adenosine 5'-monophosphate-activated protein kinase), and GCN2 (General control nonderepressible 2), which subsequently depletes epigenetic substrates feeding histone methylation at loci of a cluster of metabolism-responsive inflammatory genes, thereby suppressing their transcription. Kir2.1-mediated Vm supports nutrient uptake by facilitating cell-surface retention of nutrient transporters such as 4F2hc and GLUT1 by its modulation of plasma membrane phospholipid dynamics. Pharmacological targeting of Kir2.1 alleviated inflammation triggered by LPS or bacterial infection in a sepsis model and sterile inflammation in human samples. These findings identify an ionic control of macrophage activation and advance our understanding of the immunomodulatory properties of Vm that links nutrient inputs to inflammatory diseases.
Collapse
Affiliation(s)
- Weiwei Yu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, P. R. China
| | - Zhen Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China.,Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, P. R. China
| | - Xiafei Yu
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Yonghui Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Zili Xie
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Kailian Zhang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Zhexu Chi
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Sheng Chen
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Ting Xu
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Danlu Jiang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Xingchen Guo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Mobai Li
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Jian Zhang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Hui Fang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Dehang Yang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Yuxian Guo
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Xuyan Yang
- Department of Rheumatology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China
| | - Yingliang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, P. R. China
| | - Wei Yang
- Department of Biophysics, and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China.
| | - Di Wang
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, P. R. China. .,Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, P. R. China.
| |
Collapse
|
14
|
Tinsley SL, Allen-Petersen BL. PP2A and cancer epigenetics: a therapeutic opportunity waiting to happen. NAR Cancer 2022; 4:zcac002. [PMID: 35118387 PMCID: PMC8807117 DOI: 10.1093/narcan/zcac002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/08/2021] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
The epigenetic state of chromatin is altered by regulators which influence gene expression in response to environmental stimuli. While several post-translational modifications contribute to chromatin accessibility and transcriptional programs, our understanding of the role that specific phosphorylation sites play is limited. In cancer, kinases and phosphatases are commonly deregulated resulting in increased oncogenic signaling and loss of epigenetic regulation. Aberrant epigenetic states are known to promote cellular plasticity and the development of therapeutic resistance in many cancer types, highlighting the importance of these mechanisms to cancer cell phenotypes. Protein Phosphatase 2A (PP2A) is a heterotrimeric holoenzyme that targets a diverse array of cellular proteins. The composition of the PP2A complex influences its cellular targets and activity. For this reason, PP2A can be tumor suppressive or oncogenic depending on cellular context. Understanding the nuances of PP2A regulation and its effect on epigenetic alterations can lead to new therapeutic avenues that afford more specificity and contribute to the growth of personalized medicine in the oncology field. In this review, we summarize the known PP2A-regulated substrates and potential phosphorylation sites that contribute to cancer cell epigenetics and possible strategies to therapeutically leverage this phosphatase to suppress tumor growth.
Collapse
Affiliation(s)
- Samantha L Tinsley
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
15
|
Zhang X, Cheng L, Lu Y, Tang J, Lv Q, Chen X, Chen Y, Liu J. A MXene-Based Bionic Cascaded-Enzyme Nanoreactor for Tumor Phototherapy/Enzyme Dynamic Therapy and Hypoxia-Activated Chemotherapy. NANO-MICRO LETTERS 2021; 14:22. [PMID: 34882297 PMCID: PMC8660948 DOI: 10.1007/s40820-021-00761-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/10/2021] [Indexed: 05/05/2023]
Abstract
The enzyme-mediated elevation of reactive oxygen species (ROS) at the tumor sites has become an emerging strategy for regulating intracellular redox status for anticancer treatment. Herein, we proposed a camouflaged bionic cascaded-enzyme nanoreactor based on Ti3C2 nanosheets for combined tumor enzyme dynamic therapy (EDT), phototherapy and deoxygenation-activated chemotherapy. Briefly, glucose oxidase (GOX) and chloroperoxidase (CPO) were chemically conjugated onto Ti3C2 nanosheets, where the deoxygenation-activated drug tirapazamine (TPZ) was also loaded, and the Ti3C2-GOX-CPO/TPZ (TGCT) was embedded into nanosized cancer cell-derived membrane vesicles with high-expressed CD47 (meTGCT). Due to biomimetic membrane camouflage and CD47 overexpression, meTGCT exhibited superior immune escape and homologous targeting capacities, which could effectively enhance the tumor preferential targeting and internalization. Once internalized into tumor cells, the cascade reaction of GOX and CPO could generate HClO for efficient EDT. Simultaneously, additional laser irradiation could accelerate the enzymic-catalytic reaction rate and increase the generation of singlet oxygen (1O2). Furthermore, local hypoxia environment with the oxygen depletion by EDT would activate deoxygenation-sensitive prodrug for additional chemotherapy. Consequently, meTGCT exhibits amplified synergistic therapeutic effects of tumor phototherapy, EDT and chemotherapy for efficient tumor inhibition. This intelligent cascaded-enzyme nanoreactor provides a promising approach to achieve concurrent and significant antitumor therapy.
Collapse
Affiliation(s)
- Xiaoge Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Lili Cheng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Yao Lu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Junjie Tang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Qijun Lv
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Xiaomei Chen
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - You Chen
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong, 518107, People's Republic of China.
| |
Collapse
|
16
|
Karsai G, Steiner R, Kaech A, Lone MA, von Eckardstein A, Hornemann T. Metabolism of HSAN1- and T2DM-associated 1-deoxy-sphingolipids inhibits the migration of fibroblasts. J Lipid Res 2021; 62:100122. [PMID: 34563520 PMCID: PMC8521209 DOI: 10.1016/j.jlr.2021.100122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 11/03/2022] Open
Abstract
Hereditary sensory neuropathy type 1 (HSAN1) is a rare axonopathy, characterized by a progressive loss of sensation (pain, temperature, and vibration), neuropathic pain, and wound healing defects. HSAN1 is caused by several missense mutations in the serine palmitoyltransferase long-chain base subunit 1 and serine palmitoyltransferase long-chain base subunit 2 of the enzyme serine palmitoyltransferase-the key enzyme for the synthesis of sphingolipids. The mutations change the substrate specificity of serine palmitoyltransferase, which then forms an atypical class of 1-deoxy-sphinglipids (1-deoxySLs). Similarly, patients with type 2 diabetes mellitus also present with elevated 1-deoxySLs and a comparable clinical phenotype. The effect of 1-deoxySLs on neuronal cells was investigated in detail, but their impact on other cell types remains elusive. Here, we investigated the consequences of externally added 1-deoxySLs on the migration of fibroblasts in a scratch assay as a simplified cellular wound-healing model. We showed that 1-deoxy-sphinganine (1-deoxySA) inhibits the migration of NIH-3T3 fibroblasts in a dose- and time-dependent manner. This was not seen for a non-native, L-threo stereoisomer. Supplemented 1-deoxySA was metabolized to 1-deoxy-(dihydro)ceramide and downstream to 1-deoxy-sphingosine. Inhibiting downstream metabolism by blocking N-acylation rescued the migration phenotype. In contrast, adding 1-deoxy-sphingosine had a lesser effect on cell migration but caused the massive formation of intracellular vacuoles. Further experiments showed that the effect on cell migration was primarily mediated by 1-deoxy-dihydroceramides rather than by the free base or 1-deoxyceramides. Based on these findings, we suggest that limiting the N-acylation of 1-deoxySA could be a therapeutic approach to improve cell migration and wound healing in patients with HSAN1 and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | - Regula Steiner
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | - Andres Kaech
- Center for Microscopy and Image Analysis, University of Zürich, Zürich, Switzerland
| | - Museer A Lone
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland
| | | | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zürich, Zürich, Switzerland.
| |
Collapse
|
17
|
Liu S, Yan T, Sun J, Li F, Xu J, Sun H, Yu S, Liu J. Biomimetic Cascade Polymer Nanoreactors for Starvation and Photodynamic Cancer Therapy. Molecules 2021; 26:molecules26185609. [PMID: 34577080 PMCID: PMC8470963 DOI: 10.3390/molecules26185609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/11/2022] Open
Abstract
The selective disruption of nutritional supplements and the metabolic routes of cancer cells offer a promising opportunity for more efficient cancer therapeutics. Herein, a biomimetic cascade polymer nanoreactor (GOx/CAT-NC) was fabricated by encapsulating glucose oxidase (GOx) and catalase (CAT) in a porphyrin polymer nanocapsule for combined starvation and photodynamic anticancer therapy. Internalized by cancer cells, the GOx/CAT-NCs facilitate microenvironmental oxidation by catalyzing endogenous H2O2 to form O2, thereby accelerating intracellular glucose catabolism and enhancing cytotoxic singlet oxygen (1O2) production with infrared irradiation. The GOx/CAT-NCs have demonstrated synergistic advantages in long-term starvation therapy and powerful photodynamic therapy (PDT) in cancer treatment, which inhibits tumor cells at more than twice the rate of starvation therapy alone. The biomimetic polymer nanoreactor will further contribute to the advancement of complementary modes of spatiotemporal control of cancer therapy.
Collapse
Affiliation(s)
- Shengda Liu
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (S.L.); (T.Y.); (J.X.); (H.S.)
| | - Tengfei Yan
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (S.L.); (T.Y.); (J.X.); (H.S.)
| | - Jianxin Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China; (J.S.); (F.L.)
| | - Fei Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China; (J.S.); (F.L.)
| | - Jiayun Xu
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (S.L.); (T.Y.); (J.X.); (H.S.)
| | - Hongcheng Sun
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (S.L.); (T.Y.); (J.X.); (H.S.)
| | - Shuangjiang Yu
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (S.L.); (T.Y.); (J.X.); (H.S.)
- Correspondence: (S.Y.); (J.L.)
| | - Junqiu Liu
- Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, China; (S.L.); (T.Y.); (J.X.); (H.S.)
- Correspondence: (S.Y.); (J.L.)
| |
Collapse
|
18
|
Muley C, Bartelt A. Fuse your mitochondria, lose appetite: an anorexic, anti-obesity sphingolipid. EMBO Mol Med 2021; 13:e14618. [PMID: 34288516 PMCID: PMC8350894 DOI: 10.15252/emmm.202114618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 11/20/2022] Open
Abstract
Aberrant production of ceramides, the precursors of complex sphingolipids, is a hallmark of obesity and strongly linked to metabolic dysfunction (Meikle and Summers 2017). Ceramides are formed by recycling or de novo synthesis from sphingosine and a fatty acid side chain moiety. The side chain length determines lipotoxicity of ceramides, as those composed of C16:0 or C18:0 side chains are toxic whereas those with C24:0 or C24:1 are not (Meikle and Summers 2017). Counteracting the deleterious effects of high-fat diets (HFDs) rich in saturated fat either by inhibiting synthesis or by promoting degradation of ceramides mitigates insulin resistance and ectopic lipid accumulation (Meikle and Summers 2017). However, drugs that safely and selectively target ceramide metabolism have failed to translate into metabolic benefit in human trials so far.
Collapse
Affiliation(s)
- Carolin Muley
- Institute for Cardiovascular Prevention (IPEK)Ludwig‐Maximilians‐UniversityMunichGermany
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK)Ludwig‐Maximilians‐UniversityMunichGermany
- German Center for Cardiovascular ResearchPartner Site Munich Heart AllianceLudwig‐Maximilians University MunichMunichGermany
- Institute for Diabetes and Cancer (IDC)Helmholtz Center MunichGerman Research Center for Environmental HealthNeuherbergGermany
- Department of Molecular Metabolism & Sabri Ülker CenterHarvard T.H. Chan School of Public HealthBostonMAUSA
| |
Collapse
|
19
|
Jayashankar V, Selwan E, Hancock SE, Verlande A, Goodson MO, Eckenstein KH, Milinkeviciute G, Hoover BM, Chen B, Fleischman AG, Cramer KS, Hanessian S, Masri S, Turner N, Edinger AL. Drug-like sphingolipid SH-BC-893 opposes ceramide-induced mitochondrial fission and corrects diet-induced obesity. EMBO Mol Med 2021; 13:e13086. [PMID: 34231322 PMCID: PMC8350895 DOI: 10.15252/emmm.202013086] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023] Open
Abstract
Ceramide-induced mitochondrial fission drives high-fat diet (HFD)-induced obesity. However, molecules targeting mitochondrial dynamics have shown limited benefits in murine obesity models. Here, we reveal that these compounds are either unable to block ceramide-induced mitochondrial fission or require extended incubation periods to be effective. In contrast, targeting endolysosomal trafficking events important for mitochondrial fission rapidly and robustly prevented ceramide-induced disruptions in mitochondrial form and function. By simultaneously inhibiting ARF6- and PIKfyve-dependent trafficking events, the synthetic sphingolipid SH-BC-893 blocked palmitate- and ceramide-induced mitochondrial fission, preserved mitochondrial function, and prevented ER stress in vitro. Similar benefits were observed in the tissues of HFD-fed mice. Within 4 h of oral administration, SH-BC-893 normalized mitochondrial morphology in the livers and brains of HFD-fed mice, improved mitochondrial function in white adipose tissue, and corrected aberrant plasma leptin and adiponectin levels. As an interventional agent, SH-BC-893 restored normal body weight, glucose disposal, and hepatic lipid levels in mice consuming a HFD. In sum, the sphingolipid analog SH-BC-893 robustly and acutely blocks ceramide-induced mitochondrial dysfunction, correcting diet-induced obesity and its metabolic sequelae.
Collapse
Affiliation(s)
- Vaishali Jayashankar
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCAUSA
| | - Elizabeth Selwan
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCAUSA
| | - Sarah E Hancock
- School of Medical SciencesUniversity of New South WalesSydneyNSWAustralia
| | - Amandine Verlande
- Department of Biological ChemistryUniversity of California IrvineIrvineCAUSA
| | - Maggie O Goodson
- Department of Biological ChemistryUniversity of California IrvineIrvineCAUSA
| | - Kazumi H Eckenstein
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCAUSA
| | | | - Brianna M Hoover
- Division of Hematology/OncologyDepartment of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Bin Chen
- Department of ChemistryUniversité de MontréalMontréalQCCanada
| | - Angela G Fleischman
- Division of Hematology/OncologyDepartment of MedicineUniversity of CaliforniaIrvineCAUSA
| | - Karina S Cramer
- Department of Neurobiology and BehaviorUniversity of California IrvineIrvineCAUSA
| | | | - Selma Masri
- Department of Biological ChemistryUniversity of California IrvineIrvineCAUSA
| | - Nigel Turner
- School of Medical SciencesUniversity of New South WalesSydneyNSWAustralia
| | - Aimee L Edinger
- Department of Developmental and Cell BiologyUniversity of California IrvineIrvineCAUSA
| |
Collapse
|
20
|
Ferrarini I, Louie A, Zhou L, El-Deiry WS. ONC212 is a Novel Mitocan Acting Synergistically with Glycolysis Inhibition in Pancreatic Cancer. Mol Cancer Ther 2021; 20:1572-1583. [PMID: 34224362 DOI: 10.1158/1535-7163.mct-20-0962] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/08/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022]
Abstract
ONC212 is a fluorinated imipridone with preclinical efficacy against pancreatic and other malignancies. Although mitochondrial protease ClpP was identified as an ONC212-binding target, the mechanism leading to cancer cell death is incompletely understood. We investigated mitochondrial dysfunction and metabolic rewiring triggered by ONC212 in pancreatic cancer, a deadly malignancy with an urgent need for novel therapeutics. We found ClpP is expressed in pancreatic cancer cells and is required for ONC212 cytotoxicity. ClpX, the regulatory binding partner of ClpP, is suppressed upon ONC212 treatment. Immunoblotting and extracellular flux analysis showed ONC212 impairs oxidative phosphorylation (OXPHOS) with decrease in mitochondrial-derived ATP production. Although collapse of mitochondrial function is observed across ONC212-treated cell lines, only OXPHOS-dependent cells undergo apoptosis. Cells relying on glycolysis undergo growth arrest and upregulate glucose catabolism to prevent ERK1/2 inhibition and apoptosis. Glucose restriction or combination with glycolytic inhibitor 2-deoxy-D-glucose synergize with ONC212 and promote apoptosis in vitro and in vivo Thus, ONC212 is a novel mitocan targeting oxidative metabolism in pancreatic cancer, leading to different cellular outcomes based on divergent metabolic programs.
Collapse
Affiliation(s)
- Isacco Ferrarini
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, Rhode Island.,Department of Medicine, Section of Hematology, Cancer Research and Cell Biology Laboratory, University of Verona, Verona, Italy.,Department of Pathology and Laboratory medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island.,The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, Rhode Island.,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Anna Louie
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, Rhode Island.,Department of Pathology and Laboratory medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island.,The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, Rhode Island.,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, Rhode Island.,Department of Surgery, Brown University, Lifespan Health System and Warren, Alpert Medical School, Brown University, Providence, Rhode Island
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, Rhode Island.,Department of Pathology and Laboratory medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island.,The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, Rhode Island.,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, Rhode Island. .,Department of Pathology and Laboratory medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island.,The Joint Program in Cancer Biology, Brown University and Lifespan Health System, Providence, Rhode Island.,Cancer Center at Brown University, Warren Alpert Medical School, Brown University, Providence, Rhode Island.,Hematology-Oncology Division, Department of Medicine, Lifespan Health System and Warren Alpert Medical School, Brown University, Providence, Rhode Island
| |
Collapse
|
21
|
Haensel D, Jin S, Sun P, Cinco R, Dragan M, Nguyen Q, Cang Z, Gong Y, Vu R, MacLean AL, Kessenbrock K, Gratton E, Nie Q, Dai X. Defining Epidermal Basal Cell States during Skin Homeostasis and Wound Healing Using Single-Cell Transcriptomics. Cell Rep 2021; 30:3932-3947.e6. [PMID: 32187560 PMCID: PMC7218802 DOI: 10.1016/j.celrep.2020.02.091] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 01/07/2020] [Accepted: 02/25/2020] [Indexed: 01/17/2023] Open
Abstract
Our knowledge of transcriptional heterogeneities in epithelial stem and progenitor cell compartments is limited. Epidermal basal cells sustain cutaneous tissue maintenance and drive wound healing. Previous studies have probed basal cell heterogeneity in stem and progenitor potential, but a comprehensive dissection of basal cell dynamics during differentiation is lacking. Using single-cell RNA sequencing coupled with RNAScope and fluorescence lifetime imaging, we identify three non-proliferative and one proliferative basal cell state in homeostatic skin that differ in metabolic preference and become spatially partitioned during wound re-epithelialization. Pseudotemporal trajectory and RNA velocity analyses predict a quasi-linear differentiation hierarchy where basal cells progress from Col17a1Hi/Trp63Hi state to early-response state, proliferate at the juncture of these two states, or become growth arrested before differentiating into spinous cells. Wound healing induces plasticity manifested by dynamic basal-spinous interconversions at multiple basal transcriptional states. Our study provides a systematic view of epidermal cellular dynamics, supporting a revised “hierarchical-lineage” model of homeostasis. Haensel et al. performed a comprehensive dissection of the cellular makeup of skin during homeostasis and wound healing and the molecular heterogeneity and cellular dynamics within its stem-cell-containing epidermal basal layer. Their work provides insights and stimulates further investigation into the mechanism of skin maintenance and repair.
Collapse
Affiliation(s)
- Daniel Haensel
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- These authors contributed equally
| | - Suoqin Jin
- Department of Mathematics, University of California, Irvine, CA 92697, USA
- These authors contributed equally
| | - Peng Sun
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Rachel Cinco
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Morgan Dragan
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
| | - Quy Nguyen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Zixuan Cang
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Department of Mathematics, University of California, Irvine, CA 92697, USA
| | - Yanwen Gong
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, CA 92697, USA
| | - Remy Vu
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
| | - Adam L. MacLean
- Department of Mathematics, University of California, Irvine, CA 92697, USA
| | - Kai Kessenbrock
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA
| | - Qing Nie
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Department of Mathematics, University of California, Irvine, CA 92697, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
- Correspondence: (Q.N.), (X.D.)
| | - Xing Dai
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92627, USA
- Lead Contact
- Correspondence: (Q.N.), (X.D.)
| |
Collapse
|
22
|
Kovalenko A, Sanin A, Kosmas K, Zhang L, Wang J, Akl EW, Giannikou K, Probst CK, Hougard TR, Rue RW, Krymskaya VP, Asara JM, Lam HC, Kwiatkowski DJ, Henske EP, Filippakis H. Therapeutic Targeting of DGKA-Mediated Macropinocytosis Leads to Phospholipid Reprogramming in Tuberous Sclerosis Complex. Cancer Res 2021; 81:2086-2100. [PMID: 33593821 DOI: 10.1158/0008-5472.can-20-2218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/16/2020] [Accepted: 02/10/2021] [Indexed: 11/16/2022]
Abstract
Lymphangioleiomyomatosis is a rare destructive lung disease affecting primarily women and is the primary lung manifestation of tuberous sclerosis complex (TSC). In lymphangioleiomyomatosis, biallelic loss of TSC1/2 leads to hyperactivation of mTORC1 and inhibition of autophagy. To determine how the metabolic vulnerabilities of TSC2-deficient cells can be targeted, we performed a high-throughput screen utilizing the "Repurposing" library at the Broad Institute of MIT and Harvard (Cambridge, MA), with or without the autophagy inhibitor chloroquine. Ritanserin, an inhibitor of diacylglycerol kinase alpha (DGKA), was identified as a selective inhibitor of proliferation of Tsc2-/- mouse embryonic fibroblasts (MEF), with no impact on Tsc2+/+ MEFs. DGKA is a lipid kinase that metabolizes diacylglycerol to phosphatidic acid, a key component of plasma membranes. Phosphatidic acid levels were increased 5-fold in Tsc2-/- MEFs compared with Tsc2+/+ MEFs, and treatment of Tsc2-/- MEFs with ritanserin led to depletion of phosphatidic acid as well as rewiring of phospholipid metabolism. Macropinocytosis is known to be upregulated in TSC2-deficient cells. Ritanserin decreased macropinocytic uptake of albumin, limited the number of lysosomes, and reduced lysosomal activity in Tsc2-/- MEFs. In a mouse model of TSC, ritanserin treatment decreased cyst frequency and volume, and in a mouse model of lymphangioleiomyomatosis, genetic downregulation of DGKA prevented alveolar destruction and airspace enlargement. Collectively, these data indicate that DGKA supports macropinocytosis in TSC2-deficient cells to maintain phospholipid homeostasis and promote proliferation. Targeting macropinocytosis with ritanserin may represent a novel therapeutic approach for the treatment of TSC and lymphangioleiomyomatosis. SIGNIFICANCE: This study identifies macropinocytosis and phospholipid metabolism as novel mechanisms of metabolic homeostasis in mTORC1-hyperactive cells and suggest ritanserin as a novel therapeutic strategy for use in mTORC1-hyperactive tumors, including pancreatic cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2086/F1.large.jpg.
Collapse
Affiliation(s)
- Andrii Kovalenko
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andres Sanin
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kosmas Kosmas
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Long Zhang
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ji Wang
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elie W Akl
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Krinio Giannikou
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Clemens K Probst
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas R Hougard
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ryan W Rue
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vera P Krymskaya
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Hilaire C Lam
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David J Kwiatkowski
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth P Henske
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Harilaos Filippakis
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
23
|
Sneeggen M, Guadagno NA, Progida C. Intracellular Transport in Cancer Metabolic Reprogramming. Front Cell Dev Biol 2020; 8:597608. [PMID: 33195279 PMCID: PMC7661548 DOI: 10.3389/fcell.2020.597608] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor progression is a complex process consisting of several steps characterized by alterations in cellular behavior and morphology. These steps include uncontrolled cell division and proliferation, invasiveness and metastatic ability. Throughout these phases, cancer cells encounter a changing environment and a variety of metabolic stress. To meet their needs for energy while they proliferate and survive in their new environment, tumor cells need to continuously fine-tune their metabolism. The connection between intracellular transport and metabolic reprogramming during cancer progression is emerging as a central process of cellular adaptation to these changes. The trafficking of proteolytic enzymes, surface receptors, but also the regulation of downstream pathways, are all central to cancer progression. In this review, we summarize different hallmarks of cancer with a special focus on the role of intracellular trafficking in cell proliferation, epithelial to mesenchymal transition as well as invasion. We will further emphasize how intracellular trafficking contributes to the regulation of energy consumption and metabolism during these steps of cancer progression.
Collapse
Affiliation(s)
- Marte Sneeggen
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | - Cinzia Progida
- Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
24
|
Synthetic Sphingolipids with 1,2-Pyridazine Appendages Improve Antiproliferative Activity in Human Cancer Cell Lines. ACS Med Chem Lett 2020; 11:686-690. [PMID: 32435371 PMCID: PMC7236038 DOI: 10.1021/acsmedchemlett.9b00553] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/12/2020] [Indexed: 12/22/2022] Open
Abstract
A synthetic sphingolipid related to a ring-constrained hydroxymethyl pyrrolidine analog of FTY720 that was known to starve cancer cells to death was chemically modified to include a series of alkoxy-tethered 3,6-substituted 1,2-pyridazines. These derivatives exhibited excellent antiproliferative activity against eight human cancer cell lines from four different cancer types. A 2.5- to 9-fold reduction in IC50 in these cell lines was observed relative to the lead compound, which lacked the appended heterocycle.
Collapse
|
25
|
Kemp JA, Keebaugh A, Edson JA, Chow D, Kleinman MT, Chew YC, McCracken AN, Edinger AL, Kwon YJ. Biocompatible Chemotherapy for Leukemia by Acid-Cleavable, PEGylated FTY720. Bioconjug Chem 2020; 31:673-684. [PMID: 31986014 DOI: 10.1021/acs.bioconjchem.9b00822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Targeting the inability of cancerous cells to adapt to metabolic stress is a promising alternative to conventional cancer chemotherapy. FTY720 (Gilenya), an FDA-approved drug for the treatment of multiple sclerosis, has recently been shown to inhibit cancer progression through the down-regulation of essential nutrient transport proteins, selectively starving cancer cells to death. However, the clinical use of FTY720 for cancer therapy is prohibited because of its capability of inducing immunosuppression (lymphopenia) and bradycardia when phosphorylated upon administration. A prodrug to specifically prevent phosphorylation during circulation, hence avoiding bradycardia and lymphopenia, was synthesized by capping its hydroxyl groups with polyethylene glycol (PEG) via an acid-cleavable ketal linkage. Improved aqueous solubility was also accomplished by PEGylation. The prodrug reduces to fully potent FTY720 upon cellular uptake and induces metabolic stress in cancer cells. Enhanced release of FTY720 at a mildly acidic endosomal pH and the ability to substantially down-regulate cell-surface nutrient transporter proteins in leukemia cells only by an acid-cleaved drug were confirmed. Importantly, the prodrug demonstrated nearly identical efficacy to FTY720 in an animal model of BCR-Abl-driven leukemia without inducing bradycardia or lymphopenia in vivo, highlighting its potential clinical value. The prodrug formulation of FTY720 demonstrates the utility of precisely engineering a drug to avoid undesirable effects by tackling specific molecular mechanisms as well as a financially favorable alternative to new drug development. A multitude of existing cancer therapeutics may be explored for prodrug formulation to avoid specific side effects and preserve or enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | - Yap Ching Chew
- Zymo Research Corporation, Irvine, California 92604, United States
| | | | | | | |
Collapse
|
26
|
Blais-Lecours P, Laouafa S, Arias-Reyes C, Santos WL, Joseph V, Burgess JK, Halayko AJ, Soliz J, Marsolais D. Metabolic Adaptation of Airway Smooth Muscle Cells to an SPHK2 Substrate Precedes Cytostasis. Am J Respir Cell Mol Biol 2020; 62:35-42. [PMID: 31247144 PMCID: PMC6938129 DOI: 10.1165/rcmb.2018-0397oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 06/26/2019] [Indexed: 12/31/2022] Open
Abstract
Thickening of the airway smooth muscle is central to bronchial hyperreactivity. We have shown that the sphingosine analog (R)-2-amino-4-(4-heptyloxyphenyl)-2-methylbutanol (AAL-R) can reverse preestablished airway hyperreactivity in a chronic asthma model. Because sphingosine analogs can be metabolized by SPHK2 (sphingosine kinase 2), we investigated whether this enzyme was required for AAL-R to perturb mechanisms sustaining airway smooth muscle cell proliferation. We found that AAL-R pretreatment reduced the capacity of live airway smooth muscle cells to use oxygen for oxidative phosphorylation and increased lactate dehydrogenase activity. We also determined that SPHK2 was upregulated in airway smooth muscle cells bearing the proliferation marker Ki67 relative to their Ki67-negative counterpart. Comparing different stromal cell subsets of the lung, we found that high SPHK2 concentrations were associated with the ability of AAL-R to inhibit metabolic activity assessed by conversion of the tetrazolium dye MTT. Knockdown or pharmacological inhibition of SPHK2 reversed the effect of AAL-R on MTT conversion, indicating the essential role for this kinase in the metabolic perturbations induced by sphingosine analogs. Our results support the hypothesis that increased SPHK2 levels in proliferating airway smooth muscle cells could be exploited to counteract airway smooth muscle thickening with synthetic substrates.
Collapse
Affiliation(s)
- Pascale Blais-Lecours
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
| | - Sofien Laouafa
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
| | - Christian Arias-Reyes
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
| | - Webster L. Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia
| | - Vincent Joseph
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
- Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| | - Janette K. Burgess
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research and
- GRIAC (Groningen Research Institute for Asthma and COPD), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Andrew J. Halayko
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; and
- Biology of Breathing Group, Children’s Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Jorge Soliz
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
- Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| | - David Marsolais
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec and
- Faculty of Medicine, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
27
|
Barthelemy C, André B. Ubiquitylation and endocytosis of the human LAT1/SLC7A5 amino acid transporter. Sci Rep 2019; 9:16760. [PMID: 31728037 PMCID: PMC6856120 DOI: 10.1038/s41598-019-53065-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/22/2019] [Indexed: 12/11/2022] Open
Abstract
The human L-type amino acid transporter 1 (LAT1), also known as SLC7A5, catalyzes the transport of large neutral amino acids across the plasma membrane. As the main transporter of several essential amino acids, notably leucine, LAT1 plays an important role in mTORC1 activation. Furthermore, it is overexpressed in various types of cancer cells, where it contributes importantly to sustained growth. Despite the importance of LAT1 in normal and tumor cells, little is known about the mechanisms that might control its activity, for example by promoting its downregulation via endocytosis. Here we report that in HeLa cells, activation of protein kinase C by phorbol 12-myristate 13-acetate (PMA) triggers efficient endocytosis and degradation of LAT1. Under these conditions we found LAT1 downregulation to correlate with increased LAT1 ubiquitylation. This modification was considerably reduced in cells depleted of the Nedd4-2 ubiquitin ligase. By systematically mutagenizing the residues of the LAT1 cytosolic tails, we identified a group of three close lysines (K19, K25, K30) in the N-terminal tail that are important for PMA-induced ubiquitylation and downregulation. Our study thus unravels a mechanism of induced endocytosis of LAT1 elicited by Nedd4-2-mediated ubiquitylation of the transporter's N-terminal tail.
Collapse
Affiliation(s)
- Céline Barthelemy
- Molecular Physiology of the Cell, Université libre de Bruxelles (ULB), IBMM (Biopark), Gosselies, Belgium
| | - Bruno André
- Molecular Physiology of the Cell, Université libre de Bruxelles (ULB), IBMM (Biopark), Gosselies, Belgium.
| |
Collapse
|
28
|
FTY720 induces non-canonical phosphatidylserine externalization and cell death in acute myeloid leukemia. Cell Death Dis 2019; 10:847. [PMID: 31699964 PMCID: PMC6838108 DOI: 10.1038/s41419-019-2080-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/15/2019] [Accepted: 10/21/2019] [Indexed: 01/01/2023]
Abstract
FTY720 (fingolimod) is a FDA-approved sphingosine analog that is phosphorylated in vivo to modulate sphingosine-1-phosphate receptor (S1PR) signaling for immunosuppression in patients with refractory multiple sclerosis. FTY720 also exhibits promising anticancer efficacy in several preclinical models. While FTY720-induced cytotoxicity is not due to S1PR signaling, the mechanism remains unclear and is reported to occur through various cell death pathways. Here, we performed a systematic, mechanistic study of FTY720-induced cell death in acute myeloid leukemia (AML). We found that FTY720 induced cell death in a panel of genetically diverse AML cell lines that was accompanied by rapid phosphatidylserine (PS) externalization. Importantly, FTY720-induced PS exposure was not due to any direct effects on plasma membrane integrity and was independent of canonical signaling by regulated cell death pathways known to activate lipid flip-flop, including caspase-dependent apoptosis/pyroptosis, necroptosis, ferroptosis, and reactive oxygen species-mediated cell death. Notably, PS exposure required cellular vacuolization induced by defects in endocytic trafficking and was suppressed by the inhibition of PP2A and shedding of Annexin V-positive subcellular particles. Collectively, our studies reveal a non-canonical pathway underlying PS externalization and cell death in AML to provide mechanistic insight into the antitumor properties of FTY720.
Collapse
|
29
|
Zhang Z, Sang W, Xie L, Dai Y. Metal-organic frameworks for multimodal bioimaging and synergistic cancer chemotherapy. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.213022] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
Ikonomov OC, Sbrissa D, Shisheva A. Small molecule PIKfyve inhibitors as cancer therapeutics: Translational promises and limitations. Toxicol Appl Pharmacol 2019; 383:114771. [PMID: 31628917 DOI: 10.1016/j.taap.2019.114771] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 09/27/2019] [Accepted: 09/29/2019] [Indexed: 11/20/2022]
Abstract
Through synthesis of two rare phosphoinositides, PtdIns(3,5)P2 and PtdIns5P, the ubiquitously expressed phosphoinositide kinase PIKfyve is implicated in pleiotropic cellular functions. Small molecules specifically inhibiting PIKfyve activity cause cytoplasmic vacuolation in all dividing cells in culture yet trigger non-apoptotic death through excessive vacuolation only in cancer cells. Intriguingly, cancer cell toxicity appears to be inhibitor-specific suggesting that additional targets beyond PIKfyve are affected. One PIKfyve inhibitor - apilimod - is already in clinical trials for treatment of B-cell malignancies. However, apilimod is inactivated in cultured cells and exhibits unexpectedly low plasma levels in patients treated with maximum oral dosage. Thus, the potential widespread use of PIKfyve inhibitors as cancer therapeutics requires progress on multiple fronts: (i) advances in methods for isolating relevant cancer cells from individual patients; (ii) delineation of the molecular mechanisms potentiating the vacuolation induced by PIKfyve inhibitors in sensitive cancer cells; (iii) design of PIKfyve inhibitors with favorable pharmacokinetics; and (iv) development of effective drug combinations.
Collapse
Affiliation(s)
- Ognian C Ikonomov
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Diego Sbrissa
- Department of Urology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Assia Shisheva
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
31
|
Design, synthesis and anticancer activity of constrained sphingolipid-phenoxazine/phenothiazine hybrid constructs targeting protein phosphatase 2A. Bioorg Med Chem Lett 2019; 29:2681-2685. [PMID: 31383588 DOI: 10.1016/j.bmcl.2019.07.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/13/2022]
Abstract
Inspired by the cytotoxicity of perphenazine toward cancer cells and its ability to activate the serine/threonine protein phosphatase 2A (PP2A), we prepared series of ether-carbon linked analogs of a constrained synthetic sphingolipid analog 3, known for its cytotoxicity, nutrient transporter down-regulation and vacuolation properties, incorporating the tricyclic neuroleptics phenoxazine and phenothiazine to represent hybrid structures with possible synergistic cytotoxic activity. While the original activity of the lead compound 3 was diminished by fusion with the phenoxazine or phenothiazine tethered moieties, the corresponding 3-pyridyltetryl ether analog 10 showed cytotoxicity and nutrient transporter down-regulation similar to the lead compound 3, although it separated these PP2A-dependent phenotypes from that of vacuolation.
Collapse
|
32
|
Clark AR, Ohlmeyer M. Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration. Pharmacol Ther 2019; 201:181-201. [PMID: 31158394 PMCID: PMC6700395 DOI: 10.1016/j.pharmthera.2019.05.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric enzyme that catalyzes the selective removal of phosphate groups from protein serine and threonine residues. Emerging evidence suggests that it functions as a tumor suppressor by constraining phosphorylation-dependent signalling pathways that regulate cellular transformation and metastasis. Therefore, PP2A-activating drugs (PADs) are being actively sought and investigated as potential novel anti-cancer treatments. Here we explore the concept that PP2A also constrains inflammatory responses through its inhibitory effects on various signalling pathways, suggesting that PADs may be effective in the treatment of inflammation-mediated pathologies.
Collapse
Affiliation(s)
- Andrew R Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
| | | |
Collapse
|
33
|
Disanza A, Bisi S, Frittoli E, Malinverno C, Marchesi S, Palamidessi A, Rizvi A, Scita G. Is cell migration a selectable trait in the natural evolution of cancer development? Philos Trans R Soc Lond B Biol Sci 2019; 374:20180224. [PMID: 31431177 DOI: 10.1098/rstb.2018.0224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Selective evolutionary pressure shapes the processes and genes that enable cancer survival and expansion in a tumour-suppressive environment. A distinguishing lethal feature of malignant cancer is its dissemination and seeding of metastatic foci. A key requirement for this process is the acquisition of a migratory/invasive ability. However, how the migratory phenotype is selected for during the natural evolution of cancer and what advantage, if any, it might provide to the growing malignant cells remain open issues. In this opinion piece, we discuss three possible answers to these issues. We will examine lines of evidence from mathematical modelling of cancer evolution that indicate that migration is an intrinsic selectable property of malignant cells that directly impacts on growth dynamics and cancer geometry. Second, we will argue that migratory phenotypes can emerge as an adaptive response to unfavourable growth conditions and endow cells not only with the ability to move/invade, but also with specific metastatic traits, including drug resistance, self-renewal and survival. Finally, we will discuss the possibility that migratory phenotypes are coincidental events that emerge by happenstance in the natural evolution of cancer. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
- Andrea Disanza
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Sara Bisi
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Emanuela Frittoli
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Chiara Malinverno
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy.,Department of Oncology and Haemato-Oncology-DIPO, School of Medicine, University of Milan, Milan, Italy
| | - Stefano Marchesi
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Andrea Palamidessi
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Abrar Rizvi
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy.,Department of Oncology and Haemato-Oncology-DIPO, School of Medicine, University of Milan, Milan, Italy
| | - Giorgio Scita
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy.,Department of Oncology and Haemato-Oncology-DIPO, School of Medicine, University of Milan, Milan, Italy
| |
Collapse
|
34
|
Elgendy M, Cirò M, Hosseini A, Weiszmann J, Mazzarella L, Ferrari E, Cazzoli R, Curigliano G, DeCensi A, Bonanni B, Budillon A, Pelicci PG, Janssens V, Ogris M, Baccarini M, Lanfrancone L, Weckwerth W, Foiani M, Minucci S. Combination of Hypoglycemia and Metformin Impairs Tumor Metabolic Plasticity and Growth by Modulating the PP2A-GSK3β-MCL-1 Axis. Cancer Cell 2019; 35:798-815.e5. [PMID: 31031016 DOI: 10.1016/j.ccell.2019.03.007] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/05/2019] [Accepted: 03/27/2019] [Indexed: 01/09/2023]
Abstract
Tumor cells may adapt to metabolic challenges by alternating between glycolysis and oxidative phosphorylation (OXPHOS). To target this metabolic plasticity, we combined intermittent fasting, a clinically feasible approach to reduce glucose availability, with the OXPHOS inhibitor metformin. In mice exposed to 24-h feeding/fasting cycles, metformin impaired tumor growth only when administered during fasting-induced hypoglycemia. Synergistic anti-neoplastic effects of the metformin/hypoglycemia combination were mediated by glycogen synthase kinase 3β (GSK3β) activation downstream of PP2A, leading to a decline in the pro-survival protein MCL-1, and cell death. Mechanistically, specific activation of the PP2A-GSK3β axis was the sum of metformin-induced inhibition of CIP2A, a PP2A suppressor, and of upregulation of the PP2A regulatory subunit B56δ by low glucose, leading to an active PP2A-B56δ complex with high affinity toward GSK3β.
Collapse
Affiliation(s)
- Mohamed Elgendy
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, via Adamello 16, 20139 Milan, Italy.
| | - Marco Cirò
- Experimental Therapeutics Program, IFOM - The FIRC Institute for Molecular Oncology Foundation, Via Adamello 16, 20139 Milan, Italy
| | - Amir Hosseini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, via Adamello 16, 20139 Milan, Italy
| | - Jakob Weiszmann
- Department of Ecogenomics and Systems Biology, Faculty of Sciences, University of Vienna, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Luca Mazzarella
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, via Adamello 16, 20139 Milan, Italy
| | - Elisa Ferrari
- Experimental Therapeutics Program, IFOM - The FIRC Institute for Molecular Oncology Foundation, Via Adamello 16, 20139 Milan, Italy
| | - Riccardo Cazzoli
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, via Adamello 16, 20139 Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Andrea DeCensi
- Medical Oncology Unit, Ospedali Galliera, 16128 Genova, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Laboratori di Mercogliano, Istituto Nazionale Tumori, IRCCS-Fondazione G. Pascale, Napoli, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, via Adamello 16, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology, Università Degli Studi di Milano, Milan, Italy
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Manfred Ogris
- Laboratory of MacroMolecular Cancer Therapeutics (MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Manuela Baccarini
- Department of Microbiology, Immunobiology and Genetics, Center for Molecular Biology of the University of Vienna, Max F. Perutz Laboratories, Vienna Biocenter (VBC), 1030 Vienna, Austria
| | - Luisa Lanfrancone
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, via Adamello 16, 20139 Milan, Italy
| | - Wolfram Weckwerth
- Department of Ecogenomics and Systems Biology, Faculty of Sciences, University of Vienna, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
| | - Marco Foiani
- Experimental Therapeutics Program, IFOM - The FIRC Institute for Molecular Oncology Foundation, Via Adamello 16, 20139 Milan, Italy; Department of Oncology and Hemato-Oncology, Università Degli Studi di Milano, Milan, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, via Adamello 16, 20139 Milan, Italy; Department of Biosciences, University of Milan, 20100 Milan, Italy.
| |
Collapse
|
35
|
Ranji-Burachaloo H, Reyhani A, Gurr PA, Dunstan DE, Qiao GG. Combined Fenton and starvation therapies using hemoglobin and glucose oxidase. NANOSCALE 2019; 11:5705-5716. [PMID: 30865742 DOI: 10.1039/c8nr09107b] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Separately, Fenton and starvation cancer therapies have been recently reported as impressive methods for tumor destruction. Here, we introduce natural hemoglobin and glucose oxidase (GOx) for efficient cancer treatment following combined Fenton and starvation therapies. GOx and hemoglobin were encapsulated in zeolitic imidazolate frameworks 8 (ZIF-8) to fabricate a pH-sensitive MOF activated by tumor acidity. In the slightly acidic environment of cancer cells, GOx is released and it consumes d-glucose and molecular oxygen, nutrients essential for the survival of cancer cells, and produces gluconic acid and hydrogen peroxide, respectively. The produced gluconic acid increases the acidity of the tumor microenvironment leading to complete MOF destruction and enhances hemoglobin and GOx release. The Fe ions from the heme groups of hemoglobin also release in the presence of both endogenous and produced H2O2 and generate hydroxyl radicals. The produced OH˙ radical can rapidly oxidize the surrounding biomacromolecules in the biological system and treat the cancer cells. In vitro experiments demonstrate that this novel nanoparticle is cytotoxic to cancer cells HeLa and MCF-7, at very low concentrations (<2 μg mL-1). In addition, the selectivity index values are 5.52 and 11.04 for HeLa and MCF-7 cells, respectively, which are much higher than those of commercial drugs and those of similar studies reported by other research groups. This work thus demonstrates a novel pH-sensitive system containing hemoglobin and GOx for effective and selective cancer treatment using both radical generation and nutrient starvation.
Collapse
Affiliation(s)
- Hadi Ranji-Burachaloo
- Polymer Science Group, Department of Chemical Engineering, The University of Melbourne, Parkville, VIC 3010, Australia.
| | | | | | | | | |
Collapse
|
36
|
Kubiniok P, Finicle BT, Piffaretti F, McCracken AN, Perryman M, Hanessian S, Edinger AL, Thibault P. Dynamic Phosphoproteomics Uncovers Signaling Pathways Modulated by Anti-oncogenic Sphingolipid Analogs. Mol Cell Proteomics 2019; 18:408-422. [PMID: 30482847 PMCID: PMC6398214 DOI: 10.1074/mcp.ra118.001053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/23/2018] [Indexed: 12/31/2022] Open
Abstract
The anti-neoplastic sphingolipid analog SH-BC-893 starves cancer cells to death by down-regulating cell surface nutrient transporters and blocking lysosomal trafficking events. These effects are mediated by the activation of protein phosphatase 2A (PP2A). To identify putative PP2A substrates, we used quantitative phosphoproteomics to profile the temporal changes in protein phosphorylation in FL5.12 cells following incubation with SH-BC-893 or the specific PP2A inhibitor LB-100. These analyses enabled the profiling of more than 15,000 phosphorylation sites, of which 958 sites on 644 proteins were dynamically regulated. We identified 114 putative PP2A substrates including several nutrient transporter proteins, GTPase regulators (e.g. Agap2, Git1), and proteins associated with actin cytoskeletal remodeling (e.g. Vim, Pxn). To identify SH-BC-893-induced cell signaling events that disrupt lysosomal trafficking, we compared phosphorylation profiles in cells treated with SH-BC-893 or C2-ceramide, a non-vacuolating sphingolipid that does not impair lysosomal fusion. These analyses combined with functional assays uncovered the differential regulation of Akt and Gsk3b by SH-BC-893 (vacuolating) and C2-ceramide (non-vacuolating). Dynamic phosphoproteomics of cells treated with compounds affecting PP2A activity thus enabled the correlation of cell signaling with phenotypes to rationalize their mode of action.
Collapse
Affiliation(s)
- Peter Kubiniok
- From the ‡Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128, Succursale centre-ville, Montréal, Québec, H3C 3J7, Canada
- §Department of Chemistry, Université de Montréal, Quebec, H3C 3J7, Canada
| | - Brendan T Finicle
- ¶Department of Developmental and Cell Biology, University of California Irvine, Irvine CA 92697
| | - Fanny Piffaretti
- From the ‡Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128, Succursale centre-ville, Montréal, Québec, H3C 3J7, Canada
| | - Alison N McCracken
- ¶Department of Developmental and Cell Biology, University of California Irvine, Irvine CA 92697
| | - Michael Perryman
- §Department of Chemistry, Université de Montréal, Quebec, H3C 3J7, Canada
| | - Stephen Hanessian
- §Department of Chemistry, Université de Montréal, Quebec, H3C 3J7, Canada
| | - Aimee L Edinger
- ¶Department of Developmental and Cell Biology, University of California Irvine, Irvine CA 92697;
| | - Pierre Thibault
- From the ‡Institute for Research in Immunology and Cancer, Université de Montréal, C.P. 6128, Succursale centre-ville, Montréal, Québec, H3C 3J7, Canada;
- §Department of Chemistry, Université de Montréal, Quebec, H3C 3J7, Canada
- ‖Department of Biochemistry, Université de Montréal, C.P. 6128, Succursale centre-ville, Montréal, Québec, H3C 3J7, Canada
| |
Collapse
|
37
|
Florey O, Overholtzer M. Macropinocytosis and autophagy crosstalk in nutrient scavenging. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180154. [PMID: 30967004 PMCID: PMC6304738 DOI: 10.1098/rstb.2018.0154] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2018] [Indexed: 02/06/2023] Open
Abstract
Adaptive strategies used by cells to scavenge and recycle essential nutrients are important for survival in nutrient-depleted environments such as cancer tissues. Autophagy and macropinocytosis are two major mechanisms that promote nutrient recycling and scavenging, which share considerable, yet poorly understood, cross-regulation. Here we review recent findings that connect these starvation response mechanisms and discuss the implications of their crosstalk. This article is part of the Theo Murphy meeting issue 'Macropinocytosis'.
Collapse
Affiliation(s)
- Oliver Florey
- Signalling Programme, Babraham Institute, Cambridge CB22 3AT, UK
| | - Michael Overholtzer
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
38
|
Garsi JB, Sernissi L, Vece V, Hanessian S, McCracken AN, Simitian G, Edinger AL. In search of constrained FTY720 and phytosphingosine analogs as dual acting anticancer agents targeting metabolic and epigenetic pathways. Eur J Med Chem 2018; 159:217-242. [PMID: 30292898 PMCID: PMC6217821 DOI: 10.1016/j.ejmech.2018.09.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/13/2018] [Accepted: 09/14/2018] [Indexed: 01/04/2023]
Abstract
A series of compounds containing pyrrolidine and pyrrolizidine cores with appended hydrophobic substituents were prepared as constrained analogs of FTY720 and phytosphingosine. The effect of these compounds on the viability of cancer cells, on downregulation of the nutrient transport systems, and on their ability to cause vacuolation was studied. An attempt to inhibit HDACs with some phosphate esters of our analogs was thwarted by our failure to reproduce the reported inhibitory action of FTY720-phosphate.
Collapse
Affiliation(s)
- Jean-Baptiste Garsi
- Department of Chemistry, Université de Montréal, PO Box 6128, Station Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Lorenzo Sernissi
- Department of Chemistry, Université de Montréal, PO Box 6128, Station Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Vito Vece
- Department of Chemistry, Université de Montréal, PO Box 6128, Station Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Stephen Hanessian
- Department of Chemistry, Université de Montréal, PO Box 6128, Station Centre-Ville, Montréal, QC, H3C 3J7, Canada.
| | - Alison N McCracken
- Department of Developmental and Cell Biology, University of California, Irvine, 2128 Natural Sciences 1, CA, 92697-2300, USA
| | - Grigor Simitian
- Department of Developmental and Cell Biology, University of California, Irvine, 2128 Natural Sciences 1, CA, 92697-2300, USA
| | - Aimee L Edinger
- Department of Developmental and Cell Biology, University of California, Irvine, 2128 Natural Sciences 1, CA, 92697-2300, USA.
| |
Collapse
|
39
|
Abstract
While cancer cell proliferation depends on access to extracellular nutrients, inadequate tumour perfusion means that glucose, amino acids and lipids are often in short supply. To overcome this obstacle to growth, cancer cells utilize multiple scavenging strategies, obtaining macromolecules from the microenvironment and breaking them down in the lysosome to produce substrates for ATP generation and anabolism. Recent studies have revealed four scavenging pathways that support cancer cell proliferation in low-nutrient environments: scavenging of extracellular matrix proteins via integrins, receptor-mediated albumin uptake and catabolism, macropinocytic consumption of multiple components of the tumour microenvironment and the engulfment and degradation of entire live cells via entosis. New evidence suggests that blocking these pathways alone or in combination could provide substantial benefits to patients with incurable solid tumours. Both US Food and Drug Administration (FDA)-approved drugs and several agents in preclinical or clinical development shut down individual or multiple scavenging pathways. These therapies may increase the extent and durability of tumour growth inhibition and/or prevent the development of resistance when used in combination with existing treatments. This Review summarizes the evidence suggesting that scavenging pathways drive tumour growth, highlights recent advances that define the oncogenic signal transduction pathways that regulate scavenging and considers the benefits and detriments of therapeutic strategies targeting scavenging that are currently under development.
Collapse
Affiliation(s)
- Brendan T Finicle
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Vaishali Jayashankar
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Aimee L Edinger
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
40
|
Filippakis H, Belaid A, Siroky B, Wu C, Alesi N, Hougard T, Nijmeh J, Lam HC, Henske EP. Vps34-mediated macropinocytosis in Tuberous Sclerosis Complex 2-deficient cells supports tumorigenesis. Sci Rep 2018; 8:14161. [PMID: 30242175 PMCID: PMC6155086 DOI: 10.1038/s41598-018-32256-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/03/2018] [Indexed: 01/04/2023] Open
Abstract
Tuberous Sclerosis Complex (TSC), a rare genetic disorder with mechanistic target of rapamycin complex 1 (mTORC1) hyperactivation, is characterized by multi-organ hamartomatous benign tumors including brain, skin, kidney, and lung (Lymphangioleiomyomatosis). mTORC1 hyperactivation drives metabolic reprogramming including glucose and glutamine utilization, protein, nucleic acid and lipid synthesis. To investigate the mechanisms of exogenous nutrients uptake in Tsc2-deficient cells, we measured dextran uptake, a polysaccharide internalized via macropinocytosis. Tsc2-deficient cells showed a striking increase in dextran uptake (3-fold, p < 0.0001) relative to Tsc2-expressing cells, which was decreased (3-fold, p < 0.0001) with mTOR inhibitor, Torin1. Pharmacologic and genetic inhibition of the lipid kinase Vps34 markedly abrogated uptake of Dextran in Tsc2-deficient cells. Macropinocytosis was further increased in Tsc2-deficient cells that lack autophagic mechanisms, suggesting that autophagy inhibition leads to dependence on exogenous nutrient uptake in Tsc2-deficient cells. Treatment with a macropinocytosis inhibitor, ethylisopropylamiloride (EIPA), resulted in selective growth inhibition of Atg5-deficient, Tsc2-deficient cells (50%, p < 0.0001). Genetic inhibition of autophagy (Atg5−/− MEFs) sensitized cells with Tsc2 downregulation to the Vps34 inhibitor, SAR405, resulting in growth inhibition (75%, p < 0.0001). Finally, genetic downregulation of Vps34 inhibited tumor growth and increased tumor latency in an in vivo xenograft model of TSC. Our findings show that macropinocytosis is upregulated with Tsc2-deficiency via a Vps34-dependent mechanism to support their anabolic state. The dependence of Tsc2-deficient cells on exogenous nutrients may provide novel approaches for the treatment of TSC.
Collapse
Affiliation(s)
- Harilaos Filippakis
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Amine Belaid
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian Siroky
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Constance Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Nicola Alesi
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas Hougard
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Julie Nijmeh
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hilaire C Lam
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth P Henske
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Zobel M, Disanza A, Senic-Matuglia F, Franco M, Colaluca IN, Confalonieri S, Bisi S, Barbieri E, Caldieri G, Sigismund S, Pece S, Chavrier P, Di Fiore PP, Scita G. A NUMB-EFA6B-ARF6 recycling route controls apically restricted cell protrusions and mesenchymal motility. J Cell Biol 2018; 217:3161-3182. [PMID: 30061108 PMCID: PMC6123001 DOI: 10.1083/jcb.201802023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/17/2018] [Accepted: 06/05/2018] [Indexed: 12/13/2022] Open
Abstract
The endocytic protein NUMB has been implicated in the control of various polarized cellular processes, including the acquisition of mesenchymal migratory traits through molecular mechanisms that have only been partially defined. Here, we report that NUMB is a negative regulator of a specialized set of understudied, apically restricted, actin-based protrusions, the circular dorsal ruffles (CDRs), induced by either PDGF or HGF stimulation. Through its PTB domain, NUMB binds directly to an N-terminal NPLF motif of the ARF6 guanine nucleotide exchange factor, EFA6B, and promotes its exchange activity in vitro. In cells, a NUMB-EFA6B-ARF6 axis regulates the recycling of the actin regulatory cargo RAC1 and is critical for the formation of CDRs that mark the acquisition of a mesenchymal mode of motility. Consistently, loss of NUMB promotes HGF-induced cell migration and invasion. Thus, NUMB negatively controls membrane protrusions and the acquisition of mesenchymal migratory traits by modulating EFA6B-ARF6 activity.
Collapse
Affiliation(s)
- Martina Zobel
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Andrea Disanza
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Michel Franco
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | | | | | - Sara Bisi
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Elisa Barbieri
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Giusi Caldieri
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Sara Sigismund
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Salvatore Pece
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Philippe Chavrier
- Institut Curie, PSL Research University, Paris, France
- Centre National de la Recherche Scientifique UMR 144, Membrane and Cytoskeleton Dynamics Team, Paris, France
| | - Pier Paolo Di Fiore
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giorgio Scita
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
42
|
Chiasson-MacKenzie C, Morris ZS, Liu CH, Bradford WB, Koorman T, McClatchey AI. Merlin/ERM proteins regulate growth factor-induced macropinocytosis and receptor recycling by organizing the plasma membrane:cytoskeleton interface. Genes Dev 2018; 32:1201-1214. [PMID: 30143526 PMCID: PMC6120716 DOI: 10.1101/gad.317354.118] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/20/2018] [Indexed: 12/19/2022]
Abstract
The architectural and biochemical features of the plasma membrane are governed by its intimate association with the underlying cortical cytoskeleton. The neurofibromatosis type 2 (NF2) tumor suppressor merlin and closely related membrane:cytoskeleton-linking protein ezrin organize the membrane:cytoskeleton interface, a critical cellular compartment that both regulates and is regulated by growth factor receptors. An example of this poorly understood interrelationship is macropinocytosis, an ancient process of nutrient uptake and membrane remodeling that can both be triggered by growth factors and manage receptor availability. We show that merlin deficiency primes the membrane:cytoskeleton interface for epidermal growth factor (EGF)-induced macropinocytosis via a mechanism involving increased cortical ezrin, altered actomyosin, and stabilized cholesterol-rich membranes. These changes profoundly alter EGF receptor (EGFR) trafficking in merlin-deficient cells, favoring increased membrane levels of its heterodimerization partner, ErbB2; clathrin-independent internalization; and recycling. Our work suggests that, unlike Ras transformed cells, merlin-deficient cells do not depend on macropinocytic protein scavenging and instead exploit macropinocytosis for receptor recycling. Finally, we provide evidence that the macropinocytic proficiency of NF2-deficient cells can be used for therapeutic uptake. This work provides new insight into fundamental mechanisms of macropinocytic uptake and processing and suggests new ways to interfere with or exploit macropinocytosis in NF2 mutant and other tumors.
Collapse
Affiliation(s)
- Christine Chiasson-MacKenzie
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Zachary S Morris
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ching-Hui Liu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - William B Bradford
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Thijs Koorman
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Andrea I McClatchey
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts 02129, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
43
|
Kim SM, Nguyen TT, Ravi A, Kubiniok P, Finicle BT, Jayashankar V, Malacrida L, Hou J, Robertson J, Gao D, Chernoff J, Digman MA, Potma EO, Tromberg BJ, Thibault P, Edinger AL. PTEN Deficiency and AMPK Activation Promote Nutrient Scavenging and Anabolism in Prostate Cancer Cells. Cancer Discov 2018; 8:866-883. [PMID: 29572236 PMCID: PMC6030497 DOI: 10.1158/2159-8290.cd-17-1215] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/22/2018] [Accepted: 03/20/2018] [Indexed: 12/20/2022]
Abstract
We report that PTEN-deficient prostate cancer cells use macropinocytosis to survive and proliferate under nutrient stress. PTEN loss increased macropinocytosis only in the context of AMPK activation, revealing a general requirement for AMPK in macropinocytosis and a novel mechanism by which AMPK promotes survival under stress. In prostate cancer cells, albumin uptake did not require macropinocytosis, but necrotic cell debris proved a specific macropinocytic cargo. Isotopic labeling confirmed that macropinocytosed necrotic cell proteins fueled new protein synthesis in prostate cancer cells. Supplementation with necrotic debris, but not albumin, also maintained lipid stores, suggesting that macropinocytosis can supply nutrients other than amino acids. Nontransformed prostatic epithelial cells were not macropinocytic, but patient-derived prostate cancer organoids and xenografts and autochthonous prostate tumors all exhibited constitutive macropinocytosis, and blocking macropinocytosis limited prostate tumor growth. Macropinocytosis of extracellular material by prostate cancer cells is a previously unappreciated tumor-microenvironment interaction that could be targeted therapeutically.Significance: As PTEN-deficient prostate cancer cells proliferate in low-nutrient environments by scavenging necrotic debris and extracellular protein via macropinocytosis, blocking macropinocytosis by inhibiting AMPK, RAC1, or PI3K may have therapeutic value, particularly in necrotic tumors and in combination with therapies that cause nutrient stress. Cancer Discov; 8(7); 866-83. ©2018 AACR.See related commentary by Commisso and Debnath, p. 800This article is highlighted in the In This Issue feature, p. 781.
Collapse
Affiliation(s)
- Seong M Kim
- Department of Developmental and Cell Biology, University of California, Irvine, California
| | - Tricia T Nguyen
- Department of Developmental and Cell Biology, University of California, Irvine, California
| | - Archna Ravi
- Department of Developmental and Cell Biology, University of California, Irvine, California
| | - Peter Kubiniok
- Department of Chemistry, Université de Montréal, Quebec, Canada
| | - Brendan T Finicle
- Department of Developmental and Cell Biology, University of California, Irvine, California
| | - Vaishali Jayashankar
- Department of Developmental and Cell Biology, University of California, Irvine, California
| | - Leonel Malacrida
- Laboratory for Fluorescence Dynamics, University of California, Irvine, California
- Departamento de Fisiopatología, Hospital del Clínicas, Facultad de Medicia, Universidad de la República, Montevideo, Uruguay
| | - Jue Hou
- Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, University of California, Irvine, California
| | - Jane Robertson
- Department of Developmental and Cell Biology, University of California, Irvine, California
| | - Dong Gao
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Michelle A Digman
- Laboratory for Fluorescence Dynamics, University of California, Irvine, California
| | - Eric O Potma
- Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, University of California, Irvine, California
| | - Bruce J Tromberg
- Laser Microbeam and Medical Program, Beckman Laser Institute and Medical Clinic, University of California, Irvine, California
| | - Pierre Thibault
- Department of Chemistry, Université de Montréal, Quebec, Canada
| | - Aimee L Edinger
- Department of Developmental and Cell Biology, University of California, Irvine, California.
| |
Collapse
|
44
|
Finicle BT, Ramirez MU, Liu G, Selwan EM, McCracken AN, Yu J, Joo Y, Nguyen J, Ou K, Roy SG, Mendoza VD, Corrales DV, Edinger AL. Sphingolipids inhibit endosomal recycling of nutrient transporters by inactivating ARF6. J Cell Sci 2018; 131:jcs.213314. [PMID: 29848659 DOI: 10.1242/jcs.213314] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 05/21/2018] [Indexed: 12/15/2022] Open
Abstract
Endogenous sphingolipids (ceramide) and related synthetic molecules (FTY720, SH-BC-893) reduce nutrient access by decreasing cell surface expression of a subset of nutrient transporter proteins. Here, we report that these sphingolipids disrupt endocytic recycling by inactivating the small GTPase ARF6. Consistent with reported roles for ARF6 in maintaining the tubular recycling endosome, MICAL-L1-positive tubules were lost from sphingolipid-treated cells. We propose that ARF6 inactivation may occur downstream of PP2A activation since: (1) sphingolipids that fail to activate PP2A did not reduce ARF6-GTP levels; (2) a structurally unrelated PP2A activator disrupted tubular recycling endosome morphology and transporter localization; and (3) overexpression of a phosphomimetic mutant of the ARF6 GEF GRP1 prevented nutrient transporter loss. ARF6 inhibition alone was not toxic; however, the ARF6 inhibitors SecinH3 and NAV2729 dramatically enhanced the killing of cancer cells by SH-BC-893 without increasing toxicity to peripheral blood mononuclear cells, suggesting that ARF6 inactivation contributes to the anti-neoplastic actions of sphingolipids. Taken together, these studies provide mechanistic insight into how ceramide and sphingolipid-like molecules limit nutrient access and suppress tumor cell growth and survival.
Collapse
Affiliation(s)
- Brendan T Finicle
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Manuel U Ramirez
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Gang Liu
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Elizabeth M Selwan
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Alison N McCracken
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Jingwen Yu
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Yoosun Joo
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Jannett Nguyen
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Kevin Ou
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Saurabh Ghosh Roy
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Victor D Mendoza
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Dania Virginia Corrales
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Aimee L Edinger
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
45
|
Chintaluri K, Goulden BD, Celmenza C, Saffi G, Miraglia E, Hammond GRV, Botelho RJ. The PH domain from the Toxoplasma gondii PH-containing protein-1 (TgPH1) serves as an ectopic reporter of phosphatidylinositol 3-phosphate in mammalian cells. PLoS One 2018; 13:e0198454. [PMID: 29870544 PMCID: PMC5988325 DOI: 10.1371/journal.pone.0198454] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 05/18/2018] [Indexed: 12/29/2022] Open
Abstract
Phosphoinositide (PtdInsP) lipids recruit effector proteins to membranes to mediate a variety of functions including signal transduction and membrane trafficking. Each PtdInsP binds to a specific set of effectors through characteristic protein domains such as the PH, FYVE and PX domains. Domains with high affinity for a single PtdInsP species are useful as probes to visualize the distribution and dynamics of that PtdInsP. The endolysosomal system is governed by two primary PtdInsPs: phosphatidylinositol 3-phosphate [PtdIns(3)P] and phosphatidylinositol 3,5-bisphosphate [PtdIns(3,5)P2], which are thought to localize and control early endosomes and lysosomes/late endosomes, respectively. While PtdIns(3)P has been analysed with mammalian-derived PX and FYVE domains, PtdIns(3,5)P2 indicators remain controversial. Thus, complementary probes against these PtdInsPs are needed, including those originating from non-mammalian proteins. Here, we characterized in mammalian cells the dynamics of the PH domain from PH-containing protein-1 from the parasite Toxoplasma gondii (TgPH1), which was previously shown to bind PtdIns(3,5)P2 in vitro. However, we show that TgPH1 retains membrane-binding in PIKfyve-inhibited cells, suggesting that TgPH1 is not a viable PtdIns(3,5)P2 marker in mammalian cells. Instead, PtdIns(3)P depletion using pharmacological and enzyme-based assays dissociated TgPH1 from membranes. Indeed, TgPH1 co-localized with Rab5-positive early endosomes. In addition, TgPH1 co-localized and behaved similarly to the PX domain of p40phox and FYVE domain of EEA1, which are commonly used as PtdIns(3)P indicators. Collectively, TgPH1 offers a complementary reporter for PtdIns(3)P derived from a non-mammalian protein and that is distinct from commonly employed PX and FYVE domain-based probes.
Collapse
Affiliation(s)
- Krishna Chintaluri
- Department of Chemistry, Ryerson University, Toronto, Ontario, Canada
- The Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
| | - Brady D. Goulden
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Camilyn Celmenza
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Golam Saffi
- Department of Chemistry, Ryerson University, Toronto, Ontario, Canada
- The Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
| | - Emily Miraglia
- Department of Chemistry, Ryerson University, Toronto, Ontario, Canada
| | - Gerald R. V. Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Roberto J. Botelho
- Department of Chemistry, Ryerson University, Toronto, Ontario, Canada
- The Molecular Science Graduate Program, Ryerson University, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
46
|
Durgan J, Florey O. Cancer cell cannibalism: Multiple triggers emerge for entosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2018; 1865:831-841. [PMID: 29548938 DOI: 10.1016/j.bbamcr.2018.03.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/02/2018] [Accepted: 03/06/2018] [Indexed: 12/22/2022]
Abstract
Entosis is a form of epithelial cell engulfment and cannibalism prevalent in human cancer. Until recently, the only known trigger for entosis was loss of attachment to the extracellular matrix, as often occurs in the tumour microenvironment. However, two new studies now reveal that entosis can also occur among adherent epithelial cells, induced by mitosis or glucose starvation. Together, these findings point to the intriguing notion that certain hallmark properties of cancer cells, including anchorage independence, aberrant proliferation and metabolic stress, can converge on the induction of cell cannibalism, a phenomenon so frequently observed in tumours. In this review, we explore the molecular, cellular and biophysical mechanisms underlying entosis and discuss the impact of cell cannibalism on tumour biology.
Collapse
Affiliation(s)
- J Durgan
- Babraham Institute, Cambridge, UK.
| | - O Florey
- Babraham Institute, Cambridge, UK
| |
Collapse
|
47
|
Choy CH, Saffi G, Gray MA, Wallace C, Dayam RM, Ou ZYA, Lenk G, Puertollano R, Watkins SC, Botelho RJ. Lysosome enlargement during inhibition of the lipid kinase PIKfyve proceeds through lysosome coalescence. J Cell Sci 2018; 131:jcs.213587. [PMID: 29661845 DOI: 10.1242/jcs.213587] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/10/2018] [Indexed: 01/07/2023] Open
Abstract
Lysosomes receive and degrade cargo from endocytosis, phagocytosis and autophagy. They also play an important role in sensing and instructing cells on their metabolic state. The lipid kinase PIKfyve generates phosphatidylinositol-3,5-bisphosphate to modulate lysosome function. PIKfyve inhibition leads to impaired degradative capacity, ion dysregulation, abated autophagic flux and a massive enlargement of lysosomes. Collectively, this leads to various physiological defects, including embryonic lethality, neurodegeneration and overt inflammation. The reasons for such drastic lysosome enlargement remain unclear. Here, we examined whether biosynthesis and/or fusion-fission dynamics contribute to swelling. First, we show that PIKfyve inhibition activates TFEB, TFE3 and MITF, enhancing lysosome gene expression. However, this did not augment lysosomal protein levels during acute PIKfyve inhibition, and deletion of TFEB and/or related proteins did not impair lysosome swelling. Instead, PIKfyve inhibition led to fewer but enlarged lysosomes, suggesting that an imbalance favouring lysosome fusion over fission causes lysosome enlargement. Indeed, conditions that abated fusion curtailed lysosome swelling in PIKfyve-inhibited cells.
Collapse
Affiliation(s)
- Christopher H Choy
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3.,The Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada, M5B2K3
| | - Golam Saffi
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3.,The Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada, M5B2K3
| | - Matthew A Gray
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3
| | - Callen Wallace
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Roya M Dayam
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3.,The Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada, M5B2K3
| | - Zhen-Yi A Ou
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3
| | - Guy Lenk
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Drive, Building 50, Room 3537, Bethesda, MD 20892, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Roberto J Botelho
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada, M5B2K3 .,The Graduate Program in Molecular Science, Ryerson University, Toronto, ON, Canada, M5B2K3
| |
Collapse
|
48
|
Lima S, Takabe K, Newton J, Saurabh K, Young MM, Leopoldino AM, Hait NC, Roberts JL, Wang HG, Dent P, Milstien S, Booth L, Spiegel S. TP53 is required for BECN1- and ATG5-dependent cell death induced by sphingosine kinase 1 inhibition. Autophagy 2018; 14:942-957. [PMID: 29368980 DOI: 10.1080/15548627.2018.1429875] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The bioactive sphingolipid metabolite sphingosine-1-phosphate (S1P) and the enzyme that produces it, SPHK1 (sphingosine kinase 1), regulate many processes important for the etiology of cancer. It has been suggested that SPHK1 levels are regulated by the tumor suppressor protein TP53, a key regulator of cell cycle arrest, apoptosis, and macroautophagy/autophagy. However, little is still known of the relationship between TP53 and SPHK1 activity in the regulation of these processes. To explore this link, we examined the effects of inhibiting SPHK1 in wild-type and TP53 null cancer cell lines. SK1-I, an analog of sphingosine and isozyme-specific SPHK1 inhibitor, suppressed cancer cell growth and clonogenic survival in a TP53-dependent manner. It also more strongly enhanced intrinsic apoptosis in wild-type TP53 cells than in isogenic TP53 null cells. Intriguingly, SK1-I induced phosphorylation of TP53 on Ser15, which increases its transcriptional activity. Consequently, levels of TP53 downstream targets such as pro-apoptotic members of the BCL2 family, including BAX, BAK1, and BID were increased in wild-type but not in TP53 null cells. Inhibition of SPHK1 also increased the formation of autophagic and multivesicular bodies, and increased processing of LC3 and its localization within acidic compartments in a TP53-dependent manner. SK1-I also induced massive accumulation of vacuoles, enhanced autophagy, and increased cell death in an SPHK1-dependent manner that also required TP53 expression. Importantly, downregulation of the key regulators of autophagic flux, BECN1 and ATG5, dramatically decreased the cytotoxicity of SK1-I only in cells with TP53 expression. Hence, our results reveal that TP53 plays an important role in vacuole-associated cell death induced by SPHK1 inhibition in cancer cells.
Collapse
Affiliation(s)
- Santiago Lima
- a Department of Biochemistry and Molecular Biology , Virginia Commonwealth University School of Medicine , Richmond , VA USA
| | - Kazuaki Takabe
- a Department of Biochemistry and Molecular Biology , Virginia Commonwealth University School of Medicine , Richmond , VA USA.,c Department of Surgery and the Massey Cancer Center , Virginia Commonwealth University School of Medicine , Richmond , VA USA
| | - Jason Newton
- a Department of Biochemistry and Molecular Biology , Virginia Commonwealth University School of Medicine , Richmond , VA USA
| | - Kumar Saurabh
- a Department of Biochemistry and Molecular Biology , Virginia Commonwealth University School of Medicine , Richmond , VA USA
| | - Megan M Young
- d Department of Pharmacology , Department of Pediatrics , Penn State University College of Medicine , Hershey , PA , USA
| | - Andreia Machado Leopoldino
- b Department of Clinical Analysis, Toxicology and Food Sciences , School of Pharmaceutical Sciences of Riberião Preto , University of São Paulo , Ribeirão Preto , SP , Brazil
| | - Nitai C Hait
- a Department of Biochemistry and Molecular Biology , Virginia Commonwealth University School of Medicine , Richmond , VA USA
| | - Jane L Roberts
- a Department of Biochemistry and Molecular Biology , Virginia Commonwealth University School of Medicine , Richmond , VA USA
| | - Hong-Gang Wang
- d Department of Pharmacology , Department of Pediatrics , Penn State University College of Medicine , Hershey , PA , USA
| | - Paul Dent
- a Department of Biochemistry and Molecular Biology , Virginia Commonwealth University School of Medicine , Richmond , VA USA
| | - Sheldon Milstien
- a Department of Biochemistry and Molecular Biology , Virginia Commonwealth University School of Medicine , Richmond , VA USA
| | - Laurence Booth
- a Department of Biochemistry and Molecular Biology , Virginia Commonwealth University School of Medicine , Richmond , VA USA
| | - Sarah Spiegel
- a Department of Biochemistry and Molecular Biology , Virginia Commonwealth University School of Medicine , Richmond , VA USA
| |
Collapse
|
49
|
Abstract
Cell-intrinsic mechanisms of nutrient sensing are intimately linked to adaptive metabolic responses, and these pathways play critical roles in the complex and dynamic nutrient environment of a growing tumor. Nutrient-responsive transcription factors (e.g., HIF, SREBP, ATF4) and signaling pathways (e.g., mTORC1, AMPK) allow tumor cells to tune their metabolic output and strategies to fluctuations in nutrient availability, thus balancing tumor cell proliferation and survival with a combination of anabolic and adaptive responses. Coupling these nutrient-sensing mechanisms to the control of recycling and scavenging processes, such as autophagy and macropinocytosis, further enhances the adaptability to nutrients within tumors. Here, we discuss the key nutrient-sensing pathways active in cancer cells, how oncogenic events influence these pathways, and their likely contributions to tumor growth and survival. A better understanding of nutrient-sensing strategies and metabolic adaptations within the tumor microenvironment is critical to defining and targeting metabolic vulnerabilities in cancer.
Collapse
Affiliation(s)
- Margaret E. Torrence
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Brendan D. Manning
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
50
|
Barthelemy C, Barry AO, Twyffels L, André B. FTY720-induced endocytosis of yeast and human amino acid transporters is preceded by reduction of their inherent activity and TORC1 inhibition. Sci Rep 2017; 7:13816. [PMID: 29062000 PMCID: PMC5653847 DOI: 10.1038/s41598-017-14124-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/03/2017] [Indexed: 12/20/2022] Open
Abstract
FTY720 is a sphingoid base analog that acts as an anticancer agent in animal models. Its effect on tumor cells stems largely from its ability to trigger endocytosis of several nutrient transporters. The observation that FTY720 similarly stimulates downregulation of amino acid permeases in yeast suggests that the cellular mechanisms it targets, which are still poorly characterized, are evolutionarily conserved. We here report that adding FTY720 to yeast cells results in rapid inhibition of the intrinsic activity of multiple permeases. This effect is associated with inhibition of the TORC1 kinase complex, which in turn promotes ubiquitin-dependent permease endocytosis. Further analysis of the Gap1 permease showed that FTY720 elicits its ubiquitylation via the same factors that promote this modification when TORC1 is inhibited by rapamycin. We also show that FTY720 promotes endocytosis of the LAT1/SLC7A5 amino acid transporter in HeLa cells, this being preceded by loss of its transport activity and by mTORC1 inhibition. Our data suggest that in yeast, TORC1 deactivation resulting from FTY720-mediated inhibition of membrane transport elicits permease endocytosis. The same process seems to occur in human cells even though our data and previous reports suggest that FTY720 promotes transporter endocytosis via an additional mechanism insensitive to rapamycin.
Collapse
Affiliation(s)
- Céline Barthelemy
- Molecular Physiology of the Cell, Université libre de Bruxelles (ULB), IBMM (Biopark), Gosselies, Belgium
| | - Abdoulaye Oury Barry
- Molecular Physiology of the Cell, Université libre de Bruxelles (ULB), IBMM (Biopark), Gosselies, Belgium
| | - Laure Twyffels
- Center for Microscopy and Molecular Imaging (CMMI), Université libre de Bruxelles (ULB), IBMM (Biopark), Gosselies, Belgium
| | - Bruno André
- Molecular Physiology of the Cell, Université libre de Bruxelles (ULB), IBMM (Biopark), Gosselies, Belgium.
| |
Collapse
|