1
|
Agrawal M, Chowhan AK. Paediatric renal tumors: An insight into molecular characteristics, histomorphology and syndromic association. World J Nephrol 2025; 14:99380. [DOI: 10.5527/wjn.v14.i2.99380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/19/2024] [Accepted: 01/14/2025] [Indexed: 04/09/2025] Open
Abstract
Paediatric renal tumors are rare and accounts for about 7% of all paediatric malignant tumors. The spectrum of paediatric renal tumors ranges from benign to malignant. Benign tumors include cystic nephroma, metanephric tumors and ossifying renal tumor of infancy. Tumor with low grade malignancy includes mesoblastic nephroma. Malignant tumors are nephroblastoma, clear cell sarcoma, malignant rhabdoid tumor, anaplastic sarcoma and Ewing sarcoma. Additionally, there are molecularly defined renal tumors, which includes renal cell carcinoma (RCC) with MiT translocations, ALK-rearranged RCC, eosinophilic solid and cystic RCC and SMARCB1- deficient renal medullary carcinoma. These tumors apart from having characteristic clinical presentation and histomorphology, also carry typical molecular mutations and translocations. Certain renal tumors have association with various genetic syndromes such as Beckwith-Weidmann syndrome, Wilm’s tumor, aniridia, genitourinary anomalies and mental retardation syndrome, Denys-Drash syndrome, rhabdoid tumor predisposition syndrome and DICER syndrome. This review article focusses on molecular characteristics, histomorphology and syndromic association of pediatric renal tumors, their immunohistochemical approach to diagnosis with recent updates in molecularly defined renal tumors.
Collapse
Affiliation(s)
- Mousmi Agrawal
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences (AIIMS), Raipur 492099, Chhattisgarh, India
| | - Amit K Chowhan
- Department of Pathology and Lab Medicine, All India Institute of Medical Sciences (AIIMS), Raipur 492099, Chhattisgarh, India
| |
Collapse
|
2
|
Mladenovic A, Harik LR, Deeb KK, Genega EM, Saeed F, Gandhi JS. TFE3/TFEB altered renal cell carcinomas in end-stage renal disease setting: A single institution clinicopathological study of 4 cases. Hum Pathol 2025; 158:105783. [PMID: 40339759 DOI: 10.1016/j.humpath.2025.105783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/10/2025]
Abstract
INTRODUCTION Translocation renal cell carcinoma (tRCC) are morphologically distinct tumors having an underlying disease defining molecular alterations (commonly TFE3/TFEB gene alterations). Their occurrence in the setting of end stage renal disease (ESRD) has been rarely reported. This study was undertaken to assess the occurrence of TFE3/TFEB altered RCCs in ESRD setting at our institution. DESIGN By retrospective review, we searched our pathology database for tRCC in ESRD setting over a 14-year period. We analyzed and documented the clinical, histopathological, immunohistochemical, and molecular findings in these tumors. RESULTS Out of 223 patients of ESRD associated with RCCs, we found 4 cases of molecularly confirmed TFE3/TFEB-altered RCCs. Three of four patients were on pharmacologic immunosuppression (2 for underlying SLE and 1 for prior liver transplant). The ages ranged from 36 to 74 years (median 48 years) with an equal sex ratio. Tumors were solitary and ranged in size from 1.3 to 4.7 cm (median 2 cm). All four cases were confined to the kidney (pT1) and did not exhibit any necrosis, small vessel invasion, or sarcomatoid/rhabdoid features. The tumors exhibited characteristic morphology (solid, nested and papillary architectures with clear and eosinophilic cytoplasm in TFE3-rearranged RCCs, and biphasic morphology with basement membrane-like material in TFEB-altered RCCs). On immunohistochemistry, tumors consistently expressed cathepsin-K (3/3) & Melan-A (3/3). On molecular studies one case was confirmed via FISH study (TFEB gene rearrangement) and three cases were confirmed via RNA fusionplex (PRCC::TFE3, MED15::TFE3 and MALAT1::TFEB fusion transcripts). The median follow-up was 13 months (range 10-95 months), none of the 4 patients had any local or metastatic recurrences. One patient died of other comorbidities. Background kidney in all 4 patients exhibited variable features of ESRD. CONCLUSION TFE3/TFEB-altered RCCs are rarely encountered in ESRD. Morphological and immunohistochemical findings of tRCC in ESRD replicate those found in sporadic settings. To the best of our knowledge, our study is the first to identify TFEB-rearranged RCCs in an ESRD setting.
Collapse
Affiliation(s)
- Andrea Mladenovic
- Department of Pathology and Laboratory Medicine, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| | - Lara R Harik
- Department of Pathology and Laboratory Medicine, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| | - Kristin K Deeb
- Department of Pathology and Laboratory Medicine, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| | - Elizabeth M Genega
- Department of Pathology and Laboratory Medicine, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| | - Faisal Saeed
- Department of Pathology and Laboratory Medicine, Emory University, School of Medicine, Atlanta, GA, 30322, USA
| | - Jatin S Gandhi
- Department of Pathology and Laboratory Medicine, Emory University, School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
3
|
Albulescu A, Botezatu A, Fudulu A, Hotnog CM, Bostan M, Mihăilă M, Iancu IV, Plesa A, Brasoveanu L. Combined Effect of Conventional Chemotherapy with Epigenetic Modulators on Glioblastoma. Genes (Basel) 2025; 16:138. [PMID: 40004468 PMCID: PMC11855767 DOI: 10.3390/genes16020138] [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: 12/31/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Glioblastoma is the most common malignant primary brain tumor, characterized by necrosis, uncontrolled proliferation, infiltration, angiogenesis, apoptosis resistance, and genomic instability. Epigenetic modifiers hold promise as adjuvant therapies for gliomas, with synergistic combinations being explored to enhance efficacy and reduce toxicity. This study aimed to evaluate the effects of single or combined treatments with various anticancer drugs (Carboplatin, Paclitaxel, Avastin), natural compounds (Quercetin), and epigenetic modulators (suberoylanilide hydroxamic acid and 5-Azacytidine) on the expression of some long noncoding RNAs and methylation drivers or some functional features in the U87-MG cell line. METHODS Treated and untreated U87-MG cells were used for the evaluation of drug-induced cytotoxicity, apoptotic events, and distribution in cell cycle phases, detection of cytokine release, and assessment of gene expression and global methylation. RESULTS Cytotoxicity assays led to the selection of drug concentrations to be used in further experiments. Expression analysis revealed distinct downregulation of nearly all investigated genes and long noncoding RNAs following treatments. All treatments resulted in a higher percentage of global methylation compared to untreated controls. All treatments effectively increased levels of apoptosis, while the epigenetic modulators exhibited a lower proliferation profile, with combined treatments showing elevated values of cell lysis. CONCLUSIONS The results indicate a link between Carboplatin and Avastin treatments and DNA methylation mechanisms involving EZH2, DNMT3A, and DNMT3B, with Avastin's direct impact on these enzymes warranting further study. This research underscores the promise of platinum-based therapies combined with epigenetic drugs to reactivate silenced tumor suppressor genes and optimize methylation profiles.
Collapse
Affiliation(s)
- Adrian Albulescu
- Molecular Virology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.A.); (A.F.); (I.V.I.); (A.P.)
- Pharmacology Department, National Institute for Chemical Pharmaceutical Research and Development, 031299 Bucharest, Romania
| | - Anca Botezatu
- Molecular Virology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.A.); (A.F.); (I.V.I.); (A.P.)
| | - Alina Fudulu
- Molecular Virology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.A.); (A.F.); (I.V.I.); (A.P.)
| | - Camelia Mia Hotnog
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.M.H.); (M.B.); (M.M.); (L.B.)
| | - Marinela Bostan
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.M.H.); (M.B.); (M.M.); (L.B.)
| | - Mirela Mihăilă
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.M.H.); (M.B.); (M.M.); (L.B.)
| | - Iulia Virginia Iancu
- Molecular Virology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.A.); (A.F.); (I.V.I.); (A.P.)
| | - Adriana Plesa
- Molecular Virology Department, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (A.A.); (A.F.); (I.V.I.); (A.P.)
| | - Lorelei Brasoveanu
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.M.H.); (M.B.); (M.M.); (L.B.)
| |
Collapse
|
4
|
Tokunaga T, Hirata H, Hitaka Y, Fujii N, Kobayashi K, Hayano T, Asai Y, Shiraishi K. Role of long non‑coding RNA leucine‑rich repeat containing 75 A‑antisense RNA1 in the invasion and progression of renal cell carcinoma. Oncol Rep 2025; 53:11. [PMID: 39575481 PMCID: PMC11603548 DOI: 10.3892/or.2024.8844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/20/2024] [Indexed: 12/01/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) serve pivotal roles in cancer biology. The present study investigated the oncogenic roles of lncRNAs in renal cell carcinoma (RCC) and their potential as prognostic biomarkers. The lncRNA leucine‑rich repeat containing 75 A‑antisense RNA1 (LRRC75A‑AS1) was identified through lncRNA microarray as a potential lncRNA that may predict the efficacy of immune checkpoint inhibitor therapy and cancer progression in RCC. The present study subsequently assessed the expression of LRRC75A‑AS1 in 212 patients with clear cell RCC (ccRCC) who underwent nephrectomy, and performed in vitro functional analysis of LRRC75A‑AS1 in RCC cell lines. Additionally, the interactions between LRRC75A‑AS1, microRNA (miR)‑370‑5p and ADAMTS5 were explored. LRRC75A‑AS1 was revealed to be significantly upregulated in ccRCC tissues compared with in adjacent normal tissues, and high LRRC75A‑AS1 expression was associated with poor prognosis, including lower progression‑free survival, in patients with RCC. The knockdown of LRRC75A‑AS1 in RCC cell lines resulted in reduced cell proliferation and invasion, highlighting its role in promoting tumorigenesis. Furthermore, the interaction among LRRC75A‑AS1, miR‑370‑5p and ADAMTS5 was suggested as a regulatory mechanism underlying RCC progression. These findings indicated that LRRC75A‑AS1 may function as an oncogene in RCC, promoting cell proliferation and invasion. Its significant upregulation in ccRCC tissues and association with poor prognosis underscore its potential as a prognostic biomarker for RCC. Understanding the regulatory interactions among LRRC75A‑AS1, miR‑370‑5p and ADAMTS5 may provide new insights into the molecular mechanisms underlying RCC and facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Takanori Tokunaga
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Hiroshi Hirata
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Yukihiro Hitaka
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Nakanori Fujii
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Keita Kobayashi
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Takahide Hayano
- Department of Systems Bioinformatics, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
- The Division of Systems Medicine and Informatics, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| | - Yoshiyuki Asai
- Department of Systems Bioinformatics, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
- The Division of Systems Medicine and Informatics, Research Institute for Cell Design Medical Science, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
- AI Systems Medicine Research and Training Center, Graduate School of Medicine, Yamaguchi University and Yamaguchi University Hospital, Ube, Yamaguchi 755-8505, Japan
| | - Koji Shiraishi
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
5
|
Kanakaraj J, Chang J, Hampton LJ, Smith SC. The New WHO Category of "Molecularly Defined Renal Carcinomas": Clinical and Diagnostic Features and Management Implications. Urol Oncol 2024; 42:211-219. [PMID: 38519377 DOI: 10.1016/j.urolonc.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/27/2024] [Accepted: 02/12/2024] [Indexed: 03/24/2024]
Abstract
The evolution of classification of renal tumors has been impacted since the turn of the millennium by rapid progress in histopathology, immunohistochemistry, and molecular genetics. Together, these features have enabled firm recognition of specific, classic types of renal cell carcinomas, such as clear cell renal cell carcinoma, that in current practice trigger histologic-type specific management and treatment protocols. Now, the fifth Edition World Health Classification's new category of "Molecularly defined renal carcinomas" changes the paradigm, defining a total of seven entities based specifically on their fundamental molecular underpinnings. These tumors, which include TFE3-rearranged, TFEB-altered, ELOC-mutated, fumarate hydratase-deficient, succinate dehydrogenase-deficient, ALK-rearranged, and SMARCB1-deficient renal medullary carcinoma, encompass a wide clinical and histopathologic phenotypic spectrum of tumors. Already, important management aspects are apparent for several of these entities, while emerging therapeutic angles are coming into view. A brief, clinically-oriented introduction of the entities in this new category, focusing on relevant diagnostic, molecular, and management aspects, is the subject of this review.
Collapse
Affiliation(s)
- Jonathan Kanakaraj
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Justin Chang
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Lance J Hampton
- Division of Urology, Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA; Department of Pathology, Richmond Veterans Affairs Medical Center, Richmond, VA; VCU Massey Comprehensive Cancer Center, Richmond, VA
| | - Steven Christopher Smith
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA; Division of Urology, Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA; Department of Pathology, Richmond Veterans Affairs Medical Center, Richmond, VA; VCU Massey Comprehensive Cancer Center, Richmond, VA.
| |
Collapse
|
6
|
Sánchez-Marín D, Silva-Cázares MB, Porras-Reyes FI, García-Román R, Campos-Parra AD. Breaking paradigms: Long non-coding RNAs forming gene fusions with potential implications in cancer. Genes Dis 2024; 11:101136. [PMID: 38292185 PMCID: PMC10825296 DOI: 10.1016/j.gendis.2023.101136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/16/2023] [Accepted: 09/10/2023] [Indexed: 02/01/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) are non-coding RNAs longer than 200 nucleotides with dynamic regulatory functions. They interact with a wide range of molecules such as DNA, RNA, and proteins to modulate diverse cellular functions through several mechanisms and, if deregulated, they can lead to cancer development and progression. Recently, it has been described that lncRNAs are susceptible to form gene fusions with mRNAs or other lncRNAs, breaking the paradigm of gene fusions consisting mainly of protein-coding genes. However, their biological significance in the tumor phenotype is still uncertain. Therefore, their recent identification opens a new line of research to study their biological role in tumorigenesis, and their potential as biomarkers with clinical relevance or as therapeutic targets. The present study aimed to review the lncRNA fusions identified so far and to know which of them have been associated with a potential function. We address the current challenges to deepen their study as well as the reasons why they represent a future therapeutic window in cancer.
Collapse
Affiliation(s)
- David Sánchez-Marín
- Posgrado en Ciencias Biológicas, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, C.P. 04360, México
| | - Macrina Beatriz Silva-Cázares
- Unidad Académica Multidisciplinaria Región Altiplano, Universidad Autónoma de San Luis Potosí (UASLP), Carretera a Cedral Km 5+600, Ejido San José de la Trojes, Matehuala, San Luis Potosí, C.P. 78760, México
| | - Fany Iris Porras-Reyes
- Servicio de Anatomía Patológica, Instituto Nacional de Cancerología (INCan), Niño Jesús, Tlalpan, Ciudad de México, C.P. 14080, México
| | - Rebeca García-Román
- Instituto de Salud Pública, Universidad Veracruzana (UV), Av. Dr Luis, Dr. Castelazo Ayala s/n, Col. Industrial Ánimas, Xalapa, Veracruz, C.P. 91190, México
| | - Alma D. Campos-Parra
- Instituto de Salud Pública, Universidad Veracruzana (UV), Av. Dr Luis, Dr. Castelazo Ayala s/n, Col. Industrial Ánimas, Xalapa, Veracruz, C.P. 91190, México
| |
Collapse
|
7
|
Wu J, Cao CZ, Cui HL, Du G, Shi HZ, Liang J, Guo L, Wang YC, Zhang J, Zhou AP, Li CL, Zheng S, Shou JZ. Prognosis and Clinicopathological Characters of Adult TFEB-Altered Renal Cell Carcinoma: A Single Center Experience of 18 Cases. Clin Genitourin Cancer 2024; 22:261-268.e3. [PMID: 38104031 DOI: 10.1016/j.clgc.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION TFEB-altered renal cell carcinoma (RCC) is a rare entity characterized by the rearrangement of the TFEB gene or TFEB amplified. The therapeutic implications and long-term survival of TFEB-altered RCC remain unclear, especially for metastatic cases. MATERIALS AND METHODS The current study initially enrolled 7604 consecutive RCC patients at our center and a total of 248 patients were selected for FISH and immunohistochemistry (IHC) analysis. Eventually, eighteen TFEB-altered RCC patients were identified. We then reported the clinical, morphological, IHC, and radiological features of these cases. RESULTS The median age at initial diagnosis was 45 years, ranging from 18 years to 66 years. The majority of the TFEB-altered RCC patients were male (61.1%), with localized disease (T1-2N0M0, 77.8%). The median split TFEB fluorescent signal was 24%, ranging from 15%-80%. The morphological characteristics of TFEB-altered RCC were variable, with acinar, papillary, solid, or nest patterns. IHC and magnetic resonance imaging features of TFEB-altered RCC were nonspecific. Nine patients with localized disease received partial nephrectomy and five patients with localized disease received radical nephrectomy. During the median follow-up of 67 months, no signs of recurrence or metastasis were found in these patients. Two patients had distant metastasis and received axitinib plus PD-1 immunotherapy. One of them died at 40-month follow-up and another still alive at 88-month follow-up. CONCLUSION TFEB-altered RCC is an extremely rare variant, exhibited mixed morphological characteristics. The radiological feature lack specificity, resembling clear cell RCC or papillary RCC. Genetic analyses including FISH analysis is crucial in the diagnosis of TFEB-altered RCC. For localized TFEB-altered RCC, both radical nephrectomy and partial nephrectomy conferred satisfactory prognosis. For metastatic TFEB-altered RCC, immunotherapy-based drug combinations could be a promising treatment strategy.
Collapse
Affiliation(s)
- Jie Wu
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chuan-Zhen Cao
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
| | - Hong-Lei Cui
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gan Du
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong-Zhe Shi
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Liang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Cheng Wang
- Department of Imaging, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jin Zhang
- Department of Imaging, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ai-Ping Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chang-Ling Li
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shan Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jian-Zhong Shou
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Schmidt LS, Vocke CD, Ricketts CJ, Blake Z, Choo KK, Nielsen D, Gautam R, Crooks DR, Reynolds KL, Krolus JL, Bashyal M, Karim B, Cowen EW, Malayeri AA, Merino MJ, Srinivasan R, Ball MW, Zbar B, Marston Linehan W. PRDM10 RCC: A Birt-Hogg-Dubé-like Syndrome Associated With Lipoma and Highly Penetrant, Aggressive Renal Tumors Morphologically Resembling Type 2 Papillary Renal Cell Carcinoma. Urology 2023; 179:58-70. [PMID: 37331486 PMCID: PMC10592549 DOI: 10.1016/j.urology.2023.04.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 03/14/2023] [Accepted: 04/10/2023] [Indexed: 06/20/2023]
Abstract
OBJECTIVE To characterize the clinical manifestations and genetic basis of a familial cancer syndrome in patients with lipomas and Birt-Hogg-Dubé-like clinical manifestations including fibrofolliculomas and trichodiscomas and kidney cancer. METHODS Genomic analysis of blood and renal tumor DNA was performed. Inheritance pattern, phenotypic manifestations, and clinical and surgical management were documented. Cutaneous, subcutaneous, and renal tumor pathologic features were characterized. RESULTS Affected individuals were found to be at risk for a highly penetrant and lethal form of bilateral, multifocal papillary renal cell carcinoma. Whole genome sequencing identified a germline pathogenic variant in PRDM10 (c.2029 T>C, p.Cys677Arg), which cosegregated with disease. PRDM10 loss of heterozygosity was identified in kidney tumors. PRDM10 was predicted to abrogate expression of FLCN, a transcriptional target of PRDM10, which was confirmed by tumor expression of GPNMB, a TFE3/TFEB target and downstream biomarker of FLCN loss. In addition, a sporadic papillary RCC from the TCGA cohort was identified with a somatic PRDM10 mutation. CONCLUSION We identified a germline PRDM10 pathogenic variant in association with a highly penetrant, aggressive form of familial papillary RCC, lipomas, and fibrofolliculomas/trichodiscomas. PRDM10 loss of heterozygosity and elevated GPNMB expression in renal tumors indicate that PRDM10 alteration leads to reduced FLCN expression, driving TFE3-induced tumor formation. These findings suggest that individuals with Birt-Hogg-Dubé-like manifestations and subcutaneous lipomas, but without a germline pathogenic FLCN variant, should be screened for germline PRDM10 variants. Importantly, kidney tumors identified in patients with a pathogenic PRDM10 variant should be managed with surgical resection instead of active surveillance.
Collapse
Affiliation(s)
- Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD; Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Cathy D Vocke
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christopher J Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Zoë Blake
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Kristin K Choo
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Deborah Nielsen
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Rabindra Gautam
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Daniel R Crooks
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Krista L Reynolds
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Janis L Krolus
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Meena Bashyal
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Edward W Cowen
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD
| | - Ashkan A Malayeri
- Radiology and Imaging Sciences, Clinical Research Center, National Institutes of Health, Bethesda, MD
| | - Maria J Merino
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mark W Ball
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Berton Zbar
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
9
|
Kharouf N, Flanagan TW, Hassan SY, Shalaby H, Khabaz M, Hassan SL, Megahed M, Haikel Y, Santourlidis S, Hassan M. Tumor Microenvironment as a Therapeutic Target in Melanoma Treatment. Cancers (Basel) 2023; 15:3147. [PMID: 37370757 DOI: 10.3390/cancers15123147] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The role of the tumor microenvironment in tumor growth and therapy has recently attracted more attention in research and drug development. The ability of the microenvironment to trigger tumor maintenance, progression, and resistance is the main cause for treatment failure and tumor relapse. Accumulated evidence indicates that the maintenance and progression of tumor cells is determined by components of the microenvironment, which include stromal cells (endothelial cells, fibroblasts, mesenchymal stem cells, and immune cells), extracellular matrix (ECM), and soluble molecules (chemokines, cytokines, growth factors, and extracellular vesicles). As a solid tumor, melanoma is not only a tumor mass of monolithic tumor cells, but it also contains supporting stroma, ECM, and soluble molecules. Melanoma cells are continuously in interaction with the components of the microenvironment. In the present review, we focus on the role of the tumor microenvironment components in the modulation of tumor progression and treatment resistance as well as the impact of the tumor microenvironment as a therapeutic target in melanoma.
Collapse
Affiliation(s)
- Naji Kharouf
- Biomaterials and Bioengineering, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Unité Mixte de Recherche 1121, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
| | - Thomas W Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA
| | - Sofie-Yasmin Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Dusseldorf, Germany
| | - Hosam Shalaby
- Department of Urology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Marla Khabaz
- Department of Production, Beta Factory for Veterinary Pharmaceutical Industries, Damascus 0100, Syria
| | - Sarah-Lilly Hassan
- Department of Chemistry, Faculty of Science, Heinrich-Heine University Duesseldorf, 40225 Dusseldorf, Germany
| | - Mosaad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany
| | - Youssef Haikel
- Biomaterials and Bioengineering, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Unité Mixte de Recherche 1121, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Simeon Santourlidis
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Mohamed Hassan
- Biomaterials and Bioengineering, Institut National de la Santé et de la Recherche Médicale, Université de Strasbourg, Unité Mixte de Recherche 1121, 67000 Strasbourg, France
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Research Laboratory of Surgery-Oncology, Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
10
|
Gebrie A. Transcription factor EB as a key molecular factor in human health and its implication in diseases. SAGE Open Med 2023; 11:20503121231157209. [PMID: 36891126 PMCID: PMC9986912 DOI: 10.1177/20503121231157209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/27/2023] [Indexed: 03/07/2023] Open
Abstract
Transcription factor EB, as a component of the microphthalmia family of transcription factors, has been demonstrated to be a key controller of autophagy-lysosomal biogenesis. Transcription factor EB is activated by stressors such as nutrition and deprivation of growth factors, hypoxia, lysosomal stress, and mitochondrial injury. To achieve the ultimate functional state, it is controlled in a variety of modes, such as in its rate of transcription, post-transcriptional control, and post-translational alterations. Due to its versatile role in numerous signaling pathways, including the Wnt, calcium, AKT, and mammalian target of rapamycin complex 1 signaling pathways, transcription factor EB-originally identified to be an oncogene-is now well acknowledged as a regulator of a wide range of physiological systems, including autophagy-lysosomal biogenesis, response to stress, metabolism, and energy homeostasis. The well-known and recently identified roles of transcription factor EB suggest that this protein might play a central role in signaling networks in a number of non-communicable illnesses, such as cancer, cardiovascular disorders, drug resistance mechanisms, immunological disease, and tissue growth. The important developments in transcription factor EB research since its first description are described in this review. This review helps to advance transcription factor EB from fundamental research into therapeutic and regenerative applications by shedding light on how important a role it plays in human health and disease at the molecular level.
Collapse
Affiliation(s)
- Alemu Gebrie
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
11
|
Abstract
Ca2+ is a universal second messenger that plays a wide variety of fundamental roles in cellular physiology. Thus, to warrant selective responses and to allow rapid mobilization upon specific stimuli, Ca2+ is accumulated in organelles to keep it at very low levels in the cytoplasm during resting conditions. Major Ca2+ storage organelles include the endoplasmic reticulum (ER), the mitochondria, and as recently demonstrated, the lysosome (Xu and Ren, Annu Rev Physiol 77:57-80, 2015). The importance of Ca2+ signaling deregulation in human physiology is underscored by its involvement in several human diseases, including lysosomal storage disorders, neurodegenerative disease and cancer (Shen et al., Nat Commun 3:731, 2012; Bae et al., J Neurosci 34:11485-11503, 2014). Recent evidence strongly suggests that lysosomal Ca2+ plays a major role in the regulation of lysosomal adaptation to nutrient availability through a lysosomal signaling pathway involving the lysosomal Ca2+ channel TRPML1 and the transcription factor TFEB, a master regulator for lysosomal function and autophagy (Sardiello et al., Science 325:473-477, 2009; Settembre et al., Science 332:1429-1433, 2011; Medina et al., Nat Cell Biol 17:288-299, 2015; Di Paola et al., Cell Calcium 69:112-121, 2018). Due to the tight relationship of this lysosomal Ca2+ channel and TFEB, in this chapter, we will focus on the role of the TRPML1/TFEB pathway in the regulation of lysosomal function and autophagy.
Collapse
Affiliation(s)
- Diego Luis Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy.
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy.
| |
Collapse
|
12
|
Zhang C, Zhou L, Li S, Zhao J, Meng X, Ma L, Wang Y, Li C, Zheng L, Ming L. Obesity accelerates immune evasion of non-small cell lung carcinoma via TFEB-dependent upregulation of Siglec-15 and glycolytic reprogramming. Cancer Lett 2022; 550:215918. [PMID: 36150633 DOI: 10.1016/j.canlet.2022.215918] [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: 06/07/2022] [Revised: 08/26/2022] [Accepted: 09/09/2022] [Indexed: 11/02/2022]
Abstract
Although obesity contributes to tumor incidence and progression in various cancers, whether obesity impacts the tumor microenvironment (TME) of non-small cell lung cancer (NSCLC) remains largely under-explored. We generated NSCLC xenograft model in diet-induced obese mice and identified that TFEB is critical to accelerate obesity-related NSCLC progression with mimic intrinsic functions on tumor biology. Mechanically, TFEB binds directly to Siglec-15 promoter to upregulate Siglec-15 expression and binds to Hk2 and Ldha promoters to enhance glycolytic flux in NSCLC cells, which restrain the expansion and cytotoxic function of CD8+ T cells while maintain suppressive Treg cells in TME, jointly promoting immune evasion of NSCLC cells in obesity. Blocking tumor TFEB improves the therapeutic efficiency of anti-PD-1 in obese mice. Altogether, our data identify essential roles of TFEB in remodeling immunosuppressive TME and promoting NSCLC development in obesity, providing scientific rational for TFEB as a potential biomarker to predict immune checkpoint blockade efficiency in obese NSCLC patients.
Collapse
Affiliation(s)
- Cai Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China
| | - Lijie Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Songyang Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Junwei Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China
| | - Xianchun Meng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China
| | - Liwei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China
| | - Yongfeng Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China
| | - Cai Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Lu Zheng
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Liang Ming
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China.
| |
Collapse
|
13
|
Wang Y, Xu G, Yang H, Zhou X, Wen H. Renal Cell Carcinoma Associated with t (6;11) Translocation/TFEB Gene Fusion: A Case Report and Review of Literature. Clin Genitourin Cancer 2022; 21:309-313. [PMID: 36153295 DOI: 10.1016/j.clgc.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/16/2022] [Accepted: 08/20/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Yiming Wang
- Department of Urology, Zhejiang University Mingzhou Hospital, Ningbo, Zhejiang, China
| | - Guangjun Xu
- Department of Urology, Zhejiang University Mingzhou Hospital, Ningbo, Zhejiang, China
| | - Haitao Yang
- Department of Pathology, Zhejiang University Mingzhou Hospital, Ningbo, Zhejiang, China
| | - Xiaolong Zhou
- Department of Pathology, Zhejiang University Mingzhou Hospital, Ningbo, Zhejiang, China
| | - Haitao Wen
- Department of Urology, Zhejiang University Mingzhou Hospital, Ningbo, Zhejiang, China.
| |
Collapse
|
14
|
Tan A, Prasad R, Lee C, Jho EH. Past, present, and future perspectives of transcription factor EB (TFEB): mechanisms of regulation and association with disease. Cell Death Differ 2022; 29:1433-1449. [PMID: 35739255 PMCID: PMC9345944 DOI: 10.1038/s41418-022-01028-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 12/16/2022] Open
Abstract
Transcription factor EB (TFEB), a member of the MiT/TFE family of basic helix-loop-helix leucine zipper transcription factors, is an established central regulator of the autophagy/lysosomal-to-nucleus signaling pathway. Originally described as an oncogene, TFEB is now widely known as a regulator of various processes, such as energy homeostasis, stress response, metabolism, and autophagy-lysosomal biogenesis because of its extensive involvement in various signaling pathways, such as mTORC1, Wnt, calcium, and AKT signaling pathways. TFEB is also implicated in various human diseases, such as lysosomal storage disorders, neurodegenerative diseases, cancers, and metabolic disorders. In this review, we present an overview of the major advances in TFEB research over the past 30 years, since its description in 1990. This review also discusses the recently discovered regulatory mechanisms of TFEB and their implications for human diseases. We also summarize the moonlighting functions of TFEB and discuss future research directions and unanswered questions in the field. Overall, this review provides insight into our understanding of TFEB as a major molecular player in human health, which will take us one step closer to promoting TFEB from basic research into clinical and regenerative applications.
Collapse
Affiliation(s)
- Anderson Tan
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Renuka Prasad
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Chaerin Lee
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea
| | - Eek-Hoon Jho
- Department of Life Science, University of Seoul, Seoul, 02504, Republic of Korea.
| |
Collapse
|
15
|
Kammerer-Jacquet SF, Gandon C, Dugay F, Laguerre B, Peyronnet B, Mathieu R, Verhoest G, Bensalah K, Leroy X, Aubert S, Vermaut C, Escande F, Verkarre V, Compérat E, Ambrosetti D, Pedeutour F, Belaud-Rotureau MA, Rioux-Leclercq N. Comprehensive study of 9 novel cases of TFEB-amplified renal cell carcinoma: an aggressive tumor with frequent PDL1 expression. Histopathology 2022; 81:228-238. [PMID: 35562857 DOI: 10.1111/his.14683] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/02/2022] [Accepted: 05/12/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND & OBJECTIVES First described in 2014, renal cell carcinoma (RCC) with TFEB amplification (6p21) is a rare molecular subgroup whose diagnosis is challenging. The prognosis and therapeutic implications remain unclear. METHODS We report here the clinical, histological, immunohistochemical and genetic features of 9 novel cases. The pathological and immunohistochemical features were centrally reviewed by expert uropathologists. Fluorescence in situ hybridization (FISH) confirmed the diagnosis and comparative genomic hybridization (CGH) was performed to determine quantitative genomic alterations. We also performed an exhaustive review of the literature and compiled our data. RESULTS TFEB-amplified RCC were locally advanced with initial lymph node involvement in one case and liver metastasis in another case. They were high-grade eosinophilic tumors with papillary/pseudopapillary architecture, frequent positivity for melanocytic markers and frequent PDL1 expression. FISH demonstrated high-level TFEB amplification in 6 cases. One case showed concomitant TFEB translocation. CGH analysis identified complex alterations with frequent losses of 1p, 2q, 3p, 6p, and frequent 6p and 8q gains. VEGFA co-amplification was identified in all cases with a lower level than TFEB. The prognosis was poor with five patients having lymph node or distant metastases. CONCLUSION TFEB-amplified RCC is a rare molecular subgroup with variable morphology whose diagnosis is confirmed by FISH analysis. The complex alterations identified by CGH are consistent with an aggressive clinical behavior. The co-amplification of VEGFA and the expression of PDL1 could suggest a potential benefit from antiangiogenics and targeted immunotherapy in combination for these aggressive tumors.
Collapse
Affiliation(s)
| | - Camille Gandon
- Department of Pathology, University Hospital, Rennes, France
| | - Frederic Dugay
- UMR 6290-IGDR, Rennes, France.,Department of Cytogenetics, University Hospital, Rennes, France
| | | | | | - Romain Mathieu
- Department of Urology, University Hospital, Rennes, France
| | | | - Karim Bensalah
- Department of Urology, University Hospital, Rennes, France
| | - Xavier Leroy
- Univ.Lille, CHU Lille, Department of Pathology, F-59000, Lille, France
| | - Sebastien Aubert
- Univ.Lille, CHU Lille, Department of Pathology, F-59000, Lille, France
| | - Catherine Vermaut
- Department of Biochemistry and Molecular Biology, University Hospital, Lille, France
| | - Fabienne Escande
- Department of Biochemistry and Molecular Biology, University Hospital, Lille, France
| | - Virginie Verkarre
- Department of Pathology, HEGP, AP-HP-centre, Paris University, Paris, France
| | - Eva Compérat
- Department of Pathology, Tenon, AP-HP, Paris, France
| | | | - Florence Pedeutour
- Laboratory of Solid Tumor Genetics, University Hospital of Nice-Côte d'Azur University, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284/INSERM U1081, Nice, France
| | - Marc-Antoine Belaud-Rotureau
- UMR 6290-IGDR, Rennes, France.,Department of Cytogenetics, University Hospital, Rennes, France.,Department of Oncology, Eugène Marquis Centre, Rennes, France
| | | | | |
Collapse
|
16
|
Wei S, Testa JR, Argani P. A review of neoplasms with MITF/MiT family translocations. Histol Histopathol 2022; 37:311-321. [PMID: 35107169 DOI: 10.14670/hh-18-426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Microphthalmia-associated transcription factor (MITF/MiT) family is a group of basic helix-loop-helix leucine zipper (bHLH-LZ) transcription factors including TFE3 (TFEA), TFEB, TFEC and MITF. The first renal neoplasms involving MITF family translocation were renal cell carcinomas with chromosome translocations involving ASPL-TFE3/t(X;17)(p11.23;q25) or MALAT1-TFEB/t(6;11)(p21.1;q12), and now it is known as MiT family translocation RCC in 2016 WHO classification. Translocations involving MITF family genes also are found in other tumor types, such as perivascular epithelioid cell neoplasm (PEComa), Alveolar soft part sarcoma (ASPS), epithelioid hemangioendothelioma, ossifying fibromyxoid tumor (OFMT), and clear cell tumor with melanocytic differentiation and ACTIN-MITF translocation. In this review, we summarize the features of different types of neoplasms with MITF family translocations.
Collapse
Affiliation(s)
- Shuanzeng Wei
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Joseph R Testa
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Pedram Argani
- Department of Pathology, The Johns Hopkins University School of Medicine, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Jain V, Bose S, Arya AK, Arif T. Lysosomes in Stem Cell Quiescence: A Potential Therapeutic Target in Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:1618. [PMID: 35406389 PMCID: PMC8996909 DOI: 10.3390/cancers14071618] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
Lysosomes are cellular organelles that regulate essential biological processes such as cellular homeostasis, development, and aging. They are primarily connected to the degradation/recycling of cellular macromolecules and participate in cellular trafficking, nutritional signaling, energy metabolism, and immune regulation. Therefore, lysosomes connect cellular metabolism and signaling pathways. Lysosome's involvement in the critical biological processes has rekindled clinical interest towards this organelle for treating various diseases, including cancer. Recent research advancements have demonstrated that lysosomes also regulate the maintenance and hemostasis of hematopoietic stem cells (HSCs), which play a critical role in the progression of acute myeloid leukemia (AML) and other types of cancer. Lysosomes regulate both HSCs' metabolic networks and identity transition. AML is a lethal type of blood cancer with a poor prognosis that is particularly associated with aging. Although the genetic landscape of AML has been extensively described, only a few targeted therapies have been produced, warranting the need for further research. This review summarizes the functions and importance of targeting lysosomes in AML, while highlighting the significance of lysosomes in HSCs maintenance.
Collapse
Affiliation(s)
- Vaibhav Jain
- Abramson Cancer Center, Department of Medicine, 421 Curie Blvd., Philadelphia, PA 19104, USA;
| | - Swaroop Bose
- Department of Dermatology, Mount Sinai Icahn School of Medicine, New York, NY 10029, USA;
| | - Awadhesh K. Arya
- Department of Anesthesiology, Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Tasleem Arif
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai Icahn School of Medicine, New York, NY 10029, USA
| |
Collapse
|
18
|
Fernandez MR, Schaub FX, Yang C, Li W, Yun S, Schaub SK, Dorsey FC, Liu M, Steeves MA, Ballabio A, Tzankov A, Chen Z, Koomen JM, Berglund AE, Cleveland JL. Disrupting the MYC-TFEB Circuit Impairs Amino Acid Homeostasis and Provokes Metabolic Anergy. Cancer Res 2022; 82:1234-1250. [PMID: 35149590 DOI: 10.1158/0008-5472.can-21-1168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 12/07/2021] [Accepted: 02/08/2022] [Indexed: 11/16/2022]
Abstract
MYC family oncoproteins are regulators of metabolic reprogramming that sustains cancer cell anabolism. Normal cells adapt to nutrient-limiting conditions by activating autophagy, which is required for amino acid (AA) homeostasis. Here we report that the autophagy pathway is suppressed by Myc in normal B cells, in premalignant and neoplastic B cells of Eμ-Myc transgenic mice, and in human MYC-driven Burkitt lymphoma. Myc suppresses autophagy by antagonizing the expression and function of transcription factor EB (TFEB), a master regulator of autophagy. Mechanisms that sustained AA pools in MYC-expressing B cells include coordinated induction of the proteasome and increases in AA transport. Reactivation of the autophagy-lysosomal pathway by TFEB disabled the malignant state by disrupting mitochondrial functions, proteasome activity, amino acid transport, and amino acid and nucleotide metabolism, leading to metabolic anergy, growth arrest and apoptosis. This phenotype provides therapeutic opportunities to disable MYC-driven malignancies, including AA restriction and treatment with proteasome inhibitors.
Collapse
Affiliation(s)
- Mario R Fernandez
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute
| | - Franz X Schaub
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute
| | - Chunying Yang
- Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute
| | - Weimin Li
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute
| | | | | | | | - Min Liu
- Proteomics Core, Moffitt Cancer Center
| | | | | | | | - Zhihua Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center
| | - John M Koomen
- Department of Molecular Oncology, Moffitt Cancer Center
| | - Anders E Berglund
- Department of Biostatistics and Bioinformatics, Division of Population Sciences, H. Lee Moffitt Cancer Center & Research Institute
| | - John L Cleveland
- Department of Tumor Biology, Moffitt Cancer Center and Research Institute
| |
Collapse
|
19
|
Chao X, Wang S, Hlobik M, Ballabio A, Ni HM, Ding WX. Loss of Hepatic Transcription Factor EB Attenuates Alcohol-Associated Liver Carcinogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:87-103. [PMID: 34717896 PMCID: PMC8747011 DOI: 10.1016/j.ajpath.2021.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 09/29/2021] [Accepted: 10/07/2021] [Indexed: 01/03/2023]
Abstract
Alcohol is a well-known risk factor for hepatocellular carcinoma. Autophagy plays a dual role in liver cancer, as it suppresses tumor initiation and promotes tumor progression. Transcription factor EB (TFEB) is a master regulator of lysosomal biogenesis and autophagy, which is impaired in alcohol-related liver disease. However, the role of TFEB in alcohol-associated liver carcinogenesis is unknown. Liver-specific Tfeb knockout (KO) mice and their matched wild-type (WT) littermates were injected with the carcinogen diethylnitrosamine (DEN), followed by chronic ethanol feeding. The numbers of both total and larger tumors increased significantly in DEN-treated mice fed ethanol diet than in mice fed control diet. Although the number of tumors was not different between WT and L-Tfeb KO mice fed either control or ethanol diet, the number of larger tumors was less in L-Tfeb KO mice than in WT mice. No differences were observed in liver injury, steatosis, inflammation, ductular reaction, fibrosis, and tumor cell proliferation in DEN-treated mice fed ethanol. However, the levels of glypican 3, a marker of malignant hepatocellular carcinoma, markedly decreased in DEN-treated L-Tfeb KO mice fed ethanol in comparison to the WT mice. These findings indicate that chronic ethanol feeding promotes DEN-initiated liver tumor development, which is attenuated by genetic deletion of hepatic TFEB.
Collapse
Affiliation(s)
- Xiaojuan Chao
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Shaogui Wang
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Madeline Hlobik
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy; Medical Genetics, Department of Translational Medicine, Federico II University, Naples, Italy; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
20
|
Papke DJ, Fisch AS, Ranganathan S, O'Neill A, Breen M, Church AJ, Perez-Atayde AR, Al-Ibraheemi A. Undifferentiated Embryonal Sarcoma of the Liver With Rhabdoid Morphology Mimicking Carcinoma: Expanding the Morphologic Spectrum or a Distinct Variant? Pediatr Dev Pathol 2021; 24:564-569. [PMID: 34121507 DOI: 10.1177/10935266211018930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Undifferentiated embryonal sarcoma of the liver (UESL) is a rare aggressive neoplasm that occurs predominantly in children. Like mesenchymal hamartoma of the liver (MHL), UESL harbors recurrent rearrangements involving 19q13.3 and 19q13.4, a region of the genome that contains a primate-specific cluster of micro-RNAs. Here, we present a case of a high-grade neoplasm that arose in the left hepatic lobe of a 5-year-old male and gave rise to widespread lymph node, visceral, and soft tissue metastases. The tumor was composed of sheets, tubules, and papillae of epithelioid cells with rhabdoid morphology. INI1 and BRG1 expression were retained. Tumor cells diffusely expressed epithelial markers, including multiple keratins. While the morphologic and immunophenotypic features were suggestive of poorly differentiated carcinoma with rhabdoid features, the tumor was found to harbor the t(11;19)(q13;q13.3) translocation characteristic of UESL, as well as a TP53 mutation. Given the clinical presentation, imaging, clinical course, the tumor was classified as UESL with unusual, carcinoma-like histopathologic features. In the context of an unclassified high-grade hepatic tumor in a young child, molecular or cytogenetic testing for chromosome 19q13 alterations should be considered.
Collapse
Affiliation(s)
- David J Papke
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Adam S Fisch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Allison O'Neill
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Micheál Breen
- Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alanna J Church
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Antonio R Perez-Atayde
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alyaa Al-Ibraheemi
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Argani P. Translocation carcinomas of the kidney. Genes Chromosomes Cancer 2021; 61:219-227. [PMID: 34704642 DOI: 10.1002/gcc.23007] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/24/2021] [Indexed: 01/19/2023] Open
Abstract
The MiT subfamily of transcription factors includes TFE3, TFEB, TFEC, and MITF. Gene fusions involving two of these transcription factors have been well-characterized in renal cell carcinoma (RCC). The TFE3-rearranged RCC (also known as Xp11 translocation RCC) was first officially recognized in the 2004 World Health Organization (WHO) renal tumor classification. The TFEB-rearranged RCC, which typically harbor a t(6;11)(p21;q12) translocation which results in a MALAT1-TFEB gene fusion, were first officially recognized in the 2016 WHO renal tumor classification. These two subtypes of translocation RCC have many similarities. Both disproportionately involve young patients, although adult translocation RCC overall outnumber pediatric cases. Both often have unusual and distinctive morphologies; the TFE3-rearranged RCCs frequently have clear cells with papillary architecture and abundant psammoma bodies, while the TFEB-rearranged RCCs frequently have a biphasic appearance with both small and large epithelioid cells and nodules of basement membrane material. However, the morphology of these two neoplasms can overlap, with one mimicking the other or other more common renal neoplasms. Both of these RCC underexpress epithelial immunohistochemical markers, such as cytokeratin and epithelial membrane antigen, relative to most other RCC. Unlike other RCC, both frequently express the cysteine protease cathepsin k and often express melanocytic markers like HMB45 and Melan A. Finally, TFE3 and TFEB have overlapping functional activity as these two transcription factors frequently heterodimerize and bind to the same targets. Therefore, these two neoplasms are now grouped together under the heading of "MiT family translocation RCC." Approximately 50 renal cell carcinomas with gene fusions involving the anaplastic lymphoma kinase (ALK) gene have now been reported. While those with a Vinculin-ALK fusion have distinctive features (predilection to affect children with sickle cell trait and to show solid architecture with striking cytoplasmic vacuolization), other ALK-fusion RCCs have more varied clinical presentations and pathologic features. This review summarizes our current knowledge of these recently described RCC.
Collapse
Affiliation(s)
- Pedram Argani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Hansel DE. A 25 year perspective on advances in the pathologic assessment and diagnosis of urologic cancers. Urol Oncol 2021; 39:582-594. [PMID: 34215506 DOI: 10.1016/j.urolonc.2021.05.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/05/2021] [Accepted: 05/09/2021] [Indexed: 12/15/2022]
Abstract
Over the past 25 years, diagnostic categories in genitourinary pathology have changed dramatically. Prostate cancer reporting incorporated numerous new variant categories, recognized the importance of intraductal carcinoma, and introduced the concept of Grade Groups. Pathologic diagnosis of bladder cancer not only added new variant categories, but also modified the grading of non-invasive urothelial neoplasms and refined staging definitions. Kidney cancer classification expanded from a handful of diagnostic categories to a broad array of additional cancer types defined by unique immunohistochemical and molecular findings. Segregation of penile carcinoma by human papillomavirus status more accurately reflected pathogenesis and helped improve prediction of cancer behavior. Testicular pathology research advanced understanding of germ cell tumor subtypes and their impact on patient outcomes. Finally, adrenal gland pathology has evolved to incorporate a broader recognition of morphological variation and risk factors associated with tumor progression. Taken together, changes in pathology over the past quarter century have revolutionized our approach to genitourinary cancers. This review seeks to highlight some of the many significant changes in genitourinary pathology that have occurred during the past 25 years and emphasize impacts on clinical outcomes or therapy, as relevant.
Collapse
Affiliation(s)
- Donna E Hansel
- Department of Pathology & Laboratory Medicine, Oregon Health & Science University, Portland, OR.
| |
Collapse
|
23
|
Doronzo G, Astanina E, Bussolino F. The Oncogene Transcription Factor EB Regulates Vascular Functions. Front Physiol 2021; 12:640061. [PMID: 33912071 PMCID: PMC8072379 DOI: 10.3389/fphys.2021.640061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/17/2021] [Indexed: 12/19/2022] Open
Abstract
Transcription factor EB (TFEB) represents an emerging player in vascular biology. It belongs to the bHLH-leucine zipper transcription factor microphthalmia family, which includes microphthalmia-associated transcription factor, transcription factor E3 and transcription factor EC, and is known to be deregulated in cancer. The canonical transcriptional pathway orchestrated by TFEB adapts cells to stress in all kinds of tissues by supporting lysosomal and autophagosome biogenesis. However, emerging findings highlight that TFEB activates other genetic programs involved in cell proliferation, metabolism, inflammation and immunity. Here, we first summarize the general principles and mechanisms by which TFEB activates its transcriptional program. Then, we analyze the current knowledge of TFEB in the vascular system, placing particular emphasis on its regulatory role in angiogenesis and on the involvement of the vascular unit in inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Gabriella Doronzo
- Department of Oncology, University of Torino, Candiolo, Italy.,Laboratory of Vascular Oncology, Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Italy
| | - Elena Astanina
- Department of Oncology, University of Torino, Candiolo, Italy.,Laboratory of Vascular Oncology, Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, Candiolo, Italy.,Laboratory of Vascular Oncology, Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Italy
| |
Collapse
|
24
|
TFEB rearranged renal cell carcinoma. A clinicopathologic and molecular study of 13 cases. Tumors harboring MALAT1-TFEB, ACTB-TFEB, and the novel NEAT1-TFEB translocations constantly express PDL1. Mod Pathol 2021; 34:842-850. [PMID: 33208882 DOI: 10.1038/s41379-020-00713-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
Renal cell carcinomas with t(6;11) chromosome translocation has been classically characterized by the rearrangement of the TFEB gene, located on chromosome 6, and MALAT1 gene, located on chromosome 11. Recently, a few other genes have been described as fusion partners in TFEB rearranged renal cell carcinomas. Although most of TFEB rearranged renal cell carcinomas have an indolent behavior, in the rare cases of advanced metastatic disease targeted therapy and predictive markers remain lacking. In the present study, we collected 13 TFEB rearranged renal cell carcinomas, confirmed by FISH, analyzing their morphology and exploring the novel gene partners. Looking for predictive markers, we have also performed PDL1 immunohistochemical analysis by using four different assays (E1L3N, 22C3, SP142, and SP263). MALAT1 gene rearrangement has been found in ten tumors, five cases showing classical biphasic morphology with "rosettes", five cases without "rosettes" mimicking other renal cell carcinomas or epithelioid angiomyolipoma/pure epithelioid PEComa. We identified two different partner genes, ACTB and NEAT1, the latter previously unreported and occurring in a tumor with an unusual solid and cystic appearance. In both cases, the "rosettes" were absent. In one case no gene partner was identified. Overall, in 12 of 13 TFEB-rearranged renal cell carcinomas staining for PDL1 SP263 was observed, whereas the other antibodies were less reliable or more difficult to interpret. In conclusion, we described the third case of ACTB-TFEB rearranged renal cell carcinoma and a novel NEAT1-TFEB rearranged renal cell carcinoma, both without the distinctive biphasic morphology typical of t(6;11) renal cell carcinoma. Finally, PDL1 SP263 was constantly expressed in TFEB rearranged renal cell carcinoma with possible clinical benefit which requires further investigations.
Collapse
|
25
|
Yun S, Vincelette ND, Yu X, Watson GW, Fernandez MR, Yang C, Hitosugi T, Cheng CH, Freischel AR, Zhang L, Li W, Hou H, Schaub FX, Vedder AR, Cen L, McGraw KL, Moon J, Murphy DJ, Ballabio A, Kaufmann SH, Berglund AE, Cleveland JL. TFEB links MYC signaling to epigenetic control of myeloid differentiation and acute myeloid leukemia. Blood Cancer Discov 2021; 2:162-185. [PMID: 33860275 PMCID: PMC8043621 DOI: 10.1158/2643-3230.bcd-20-0029] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 10/30/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
MYC oncoproteins regulate transcription of genes directing cell proliferation, metabolism and tumorigenesis. A variety of alterations drive MYC expression in acute myeloid leukemia (AML) and enforced MYC expression in hematopoietic progenitors is sufficient to induce AML. Here we report that AML and myeloid progenitor cell growth and survival rely on MYC-directed suppression of Transcription Factor EB (TFEB), a master regulator of the autophagy-lysosome pathway. Notably, although originally identified as an oncogene, TFEB functions as a tumor suppressor in AML, where it provokes AML cell differentiation and death. These responses reflect TFEB control of myeloid epigenetic programs, by inducing expression of isocitrate dehydrogenase-1 (IDH1) and IDH2, resulting in global hydroxylation of 5-methycytosine. Finally, activating the TFEB-IDH1/IDH2-TET2 axis is revealed as a targetable vulnerability in AML. Thus, epigenetic control by a MYC-TFEB circuit dictates myeloid cell fate and is essential for maintenance of AML.
Collapse
Affiliation(s)
- Seongseok Yun
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nicole D Vincelette
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Xiaoqing Yu
- Department of Bioinformatics and Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Gregory W Watson
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mario R Fernandez
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Chunying Yang
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Taro Hitosugi
- Department of Molecular Pharmacology and Experimental Therapeutics, and Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Chia-Ho Cheng
- Department of Bioinformatics and Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Audrey R Freischel
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Ling Zhang
- Department of Pathology and Laboratory Medicine, Tampa, Florida
| | - Weimin Li
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hsinan Hou
- Department of Internal Medicine, National Taiwan University, Taipei, Taiwan
| | - Franz X Schaub
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Alexis R Vedder
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Ling Cen
- Department of Bioinformatics and Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kathy L McGraw
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jungwon Moon
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Daniel J Murphy
- University of Glasgow, Institute of Cancer Sciences, Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, United Kingdom
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas
- SSM School for Advanced Studies, Federico II University, Naples, Italy
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, and Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Anders E Berglund
- Department of Bioinformatics and Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - John L Cleveland
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| |
Collapse
|
26
|
Vangoor VR, Gomes‐Duarte A, Pasterkamp RJ. Long non-coding RNAs in motor neuron development and disease. J Neurochem 2021; 156:777-801. [PMID: 32970857 PMCID: PMC8048821 DOI: 10.1111/jnc.15198] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022]
Abstract
Long non-coding RNAs (lncRNAs) are RNAs that exceed 200 nucleotides in length and that are not translated into proteins. Thousands of lncRNAs have been identified with functions in processes such as transcription and translation regulation, RNA processing, and RNA and protein sponging. LncRNAs show prominent expression in the nervous system and have been implicated in neural development, function and disease. Recent work has begun to report on the expression and roles of lncRNAs in motor neurons (MNs). The cell bodies of MNs are located in cortex, brainstem or spinal cord and their axons project into the brainstem, spinal cord or towards peripheral muscles, thereby controlling important functions such as movement, breathing and swallowing. Degeneration of MNs is a pathological hallmark of diseases such as amyotrophic lateral sclerosis and spinal muscular atrophy. LncRNAs influence several aspects of MN development and disruptions in these lncRNA-mediated effects are proposed to contribute to the pathogenic mechanisms underlying MN diseases (MNDs). Accumulating evidence suggests that lncRNAs may comprise valuable therapeutic targets for different MNDs. In this review, we discuss the role of lncRNAs (including circular RNAs [circRNAs]) in the development of MNs, discuss how lncRNAs may contribute to MNDs and provide directions for future research.
Collapse
Affiliation(s)
- Vamshidhar R. Vangoor
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain CenterUtrecht UniversityUtrechtThe Netherlands
| | - Andreia Gomes‐Duarte
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain CenterUtrecht UniversityUtrechtThe Netherlands
| | - R. Jeroen Pasterkamp
- Department of Translational NeuroscienceUniversity Medical Center Utrecht Brain CenterUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
27
|
Lu H, Sun J, Hamblin MH, Chen YE, Fan Y. Transcription factor EB regulates cardiovascular homeostasis. EBioMedicine 2021; 63:103207. [PMID: 33418500 PMCID: PMC7804971 DOI: 10.1016/j.ebiom.2020.103207] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/11/2020] [Accepted: 12/28/2020] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death and a major cause of disability globally. Transcription factor EB (TFEB), as a member of the microphthalmia transcription factor (MITF) family, has been demonstrated to be a master regulator of autophagy and lysosomal biogenesis. Emerging studies suggest that TFEB regulates homeostasis in the cardiovascular system and shows beneficial effects on CVDs, including atherosclerosis, aortic aneurysm, postischemic angiogenesis, and cardiotoxicity, constituting a promising molecular target for the prevention and treatment of these diseases. Post-translational modifications regulate TFEB nuclear translocation and its transcriptional activity. Therapeutic strategies have been pursued to enhance TFEB activity and facilitate TFEB beneficial effects on CVDs. The elucidation of TFEB function and the precise underlying mechanisms will accelerate drug development and potential applications of TFEB drugs in the treatment of human diseases.
Collapse
Affiliation(s)
- Haocheng Lu
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Jinjian Sun
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Yanbo Fan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA; Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
28
|
Theodosakis N, Pagan AD, Fisher DE. The role of MiT/TFE family members in autophagy regulation. CURRENT TOPICS IN BIOCHEMICAL RESEARCH 2021; 22:151-159. [PMID: 35663368 PMCID: PMC9165699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The MiT/TFE family of proteins are important regulators of a number of metabolic processes. One of their most important roles is activating the autophagy pathway in the setting of nutrient deprivation or buildup of toxic metabolites. Their proper and improper functioning in this role has been linked to several types of disease, including cancer and multiple forms of neurodegeneration. In this review we will briefly outline what is known about individual family members' roles in regulating autophagy across a variety of contexts.
Collapse
Affiliation(s)
| | - Angel D Pagan
- Department of Dermatology, Massachusetts General Hospital, Boston, MA, USA
- Ponce Health Sciences University School of Medicine, Ponce, Puerto Rico, USA
| | - David E Fisher
- Department of Dermatology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
29
|
Astanina E, Bussolino F, Doronzo G. Multifaceted activities of transcription factor EB in cancer onset and progression. Mol Oncol 2020; 15:327-346. [PMID: 33252196 PMCID: PMC7858119 DOI: 10.1002/1878-0261.12867] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/11/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Transcription factor EB (TFEB) represents an emerging player in cancer biology. Together with microphthalmia‐associated transcription factor, transcription factor E3 and transcription factor EC, TFEB belongs to the microphthalmia family of bHLH‐leucine zipper transcription factors that may be implicated in human melanomas, renal and pancreatic cancers. TFEB was originally described as being translocated in a juvenile subset of pediatric renal cell carcinoma; however, whole‐genome sequencing reported that somatic mutations were sporadically found in many different cancers. Besides its oncogenic activity, TFEB controls the autophagy‐lysosomal pathway by recognizing a recurrent motif present in the promoter regions of a set of genes that participate in lysosome biogenesis; furthermore, its dysregulation was found to have a crucial pathogenic role in different tumors by modulating the autophagy process. Other than regulating cancer cell‐autonomous responses, recent findings indicate that TFEB participates in the regulation of cellular functions of the tumor microenvironment. Here, we review the emerging role of TFEB in regulating cancer cell behavior and choreographing tumor–microenvironment interaction. Recognizing TFEB as a hub of network of signals exchanged within the tumor between cancer and stroma cells provides a fresh perspective on the molecular principles of tumor self‐organization, promising to reveal numerous new and potentially druggable vulnerabilities.
Collapse
Affiliation(s)
- Elena Astanina
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Italy
| | - Gabriella Doronzo
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute-IRCCS-FPO, Candiolo, Italy
| |
Collapse
|
30
|
da Costa A, Metais T, Mouthon F, Kerkovich D, Charvériat M. Evaluating and modulating TFEB in the control of autophagy: toward new treatments in CNS disorders. Fundam Clin Pharmacol 2020; 35:539-551. [PMID: 33259088 DOI: 10.1111/fcp.12634] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/15/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022]
Abstract
TFEB is a mammalian transcription factor that binds directly to the CLEAR consensus sequence (5'-GTCACGTGAC-3') present in the regulatory regions of genes inducing autophagosome formation, autophagosome-lysosome fusion, hydrolase enzyme expression, and lysosomal exocytosis. By modulating these activities, TFEB coordinates on-demand control over each cell's degradation pathway. Thus, a nuclear signaling pathway regulates cellular energy metabolism through TFEB. Our growing understanding of the role of TFEB and CLEAR in the promotion of healthy clearance together with in vitro and in vivo preclinical findings in various animal models of disease supports the conclusion that the pharmacological activation of TFEB could clear toxic proteins to treat both rare and common forms of neurodegenerative disease.
Collapse
|
31
|
Eble JN. Contributions of genetics to the evolution of the diagnostic classification of renal cell neoplasia: a personal perspective. Pathology 2020; 53:96-100. [PMID: 33234231 DOI: 10.1016/j.pathol.2020.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/22/2020] [Indexed: 12/16/2022]
Abstract
The classification system for neoplasms of the cells lining the renal tubules (renal cell neoplasms) has expanded greatly over the last five decades. The criteria for recognising an entity and including it in the classification have changed from being purely morphological and clinical to include genetics; presently, some are defined purely on genetics. Expansion of the number of entities included in the classification has many of the newly included entities and those under consideration for inclusion being very rare. The clinical utility of including entities which are extremely rare, based mainly upon genetic information, is unclear.
Collapse
Affiliation(s)
- John N Eble
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
32
|
Pederzoli F, Bandini M, Marandino L, Ali SM, Madison R, Chung J, Ross JS, Necchi A. Targetable gene fusions and aberrations in genitourinary oncology. Nat Rev Urol 2020; 17:613-625. [PMID: 33046892 DOI: 10.1038/s41585-020-00379-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
Gene fusions result from either structural chromosomal rearrangement or aberrations caused by splicing or transcriptional readthrough. The precise and distinctive presence of fusion genes in neoplastic tissues and their involvement in multiple pathways central to cancer development, growth and survival make them promising targets for personalized therapy. In genitourinary malignancies, rearrangements involving the E26 transformation-specific family of transcription factors have emerged as very frequent alterations in prostate cancer, especially the TMPRSS2-ERG fusion. In renal malignancies, Xp11 and t(6;11) translocations are hallmarks of a distinct pathological group of tumours described as microphthalmia-associated transcription factor family translocation-associated renal cell carcinomas. Novel druggable fusion events have been recognized in genitourinary malignancies, leading to the activation of several clinical trials. For instance, ALK-rearranged renal cell carcinomas have shown responses to alectinib and crizotinib. Erdafitinib has been tested for the treatment of FGFR-rearranged bladder cancer. Other anti-fibroblast growth factor receptor 3 (FGFR3) compounds are showing promising results in the treatment of bladder cancer, including infigratinib and pemigatinib, and all are currently in clinical trials.
Collapse
Affiliation(s)
- Filippo Pederzoli
- Urological Research Institute (URI), Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy.
| | - Marco Bandini
- Urological Research Institute (URI), Unit of Urology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Laura Marandino
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Siraj M Ali
- Foundation Medicine Inc., Cambridge, MA, USA
| | | | - Jon Chung
- Foundation Medicine Inc., Cambridge, MA, USA
| | - Jeffrey S Ross
- Foundation Medicine Inc., Cambridge, MA, USA.,Upstate Medical University, Syracuse, NY, USA
| | - Andrea Necchi
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
33
|
The anti-tumor agent, Dp44mT, promotes nuclear translocation of TFEB via inhibition of the AMPK-mTORC1 axis. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165970. [PMID: 32950675 DOI: 10.1016/j.bbadis.2020.165970] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022]
Abstract
Di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and its analogues are potent anti-cancer agents through their ability to target lysosomes. Considering this, it was important to understand the mechanisms involved in the Dp44mT-mediated induction of autophagy and the role of 5'-adenosine monophosphate-activated protein kinase (AMPK) as a critical autophagic regulator. As such, this investigation examined AMPK's role in the regulation of the transcription factor EB (TFEB), which transcribes genes involved in autophagy and lysosome biosynthesis. For the first time, this study demonstrated that Dp44mT induces translocation of TFEB to the nucleus. Furthermore, Dp44mT-mediated nuclear translocation of TFEB was AMPK-dependent. Considering that: (1) the mammalian target of rapamycin complex 1 (mTORC1) plays an important role in the regulation of TFEB; and (2) that AMPK is a known regulator of mTORC1, this study also elucidated the mechanisms through which Dp44mT regulates nuclear translocation of TFEB via AMPK. Silencing AMPK led to increased mTOR phosphorylation, that activates mTORC1. Since Dp44mT inhibits mTORC1 in an AMPK-dependent manner through raptor phosphorylation, Dp44mT is demonstrated to regulate TFEB translocation through dual mechanisms: AMPK activation, which inhibits mTOR, and inhibition of mTORC1 via phosphorylation of raptor. Collectively, Dp44mT-mediated activation of AMPK plays a crucial role in lysosomal biogenesis and TFEB function. As Dp44mT potently chelates copper and iron that are crucial for tumor growth, these studies provide insight into the regulatory mechanisms involved in intracellular clearance and energy metabolism that occur upon alterations in metal ion homeostasis.
Collapse
|
34
|
Vu HN, Dilshat R, Fock V, Steingrímsson E. User guide to MiT-TFE isoforms and post-translational modifications. Pigment Cell Melanoma Res 2020; 34:13-27. [PMID: 32846025 DOI: 10.1111/pcmr.12922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/31/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022]
Abstract
The microphthalmia-associated transcription factor (MITF) is at the core of melanocyte and melanoma fate specification. The related factors TFEB and TFE3 have been shown to be instrumental for transcriptional regulation of genes involved in lysosome biogenesis and autophagy, cellular processes important for mediating nutrition signals and recycling of cellular materials, in many cell types. The MITF, TFEB, TFE3, and TFEC proteins are highly related. They share many structural and functional features and are targeted by the same signaling pathways. However, the existence of several isoforms of each factor and the increasing number of residues shown to be post-translationally modified by various signaling pathways poses a difficulty in indexing amino acid residues in different isoforms across the different proteins. Here, we provide a resource manual to cross-reference amino acids and post-translational modifications in all isoforms of the MiT-TFE family in humans, mice, and zebrafish and summarize the protein accession numbers for each isoform of these factors in the different genomic databases. This will facilitate future studies on the signaling pathways that regulate different isoforms of the MiT-TFE transcription factor family.
Collapse
Affiliation(s)
- Hong Nhung Vu
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Ramile Dilshat
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Valerie Fock
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Eiríkur Steingrímsson
- Department of Biochemistry and Molecular Biology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| |
Collapse
|
35
|
Montella M, Franco R, Aquino G, Ronchi A, Zito Marino F, Di Napoli M, Pignata S, Cozzolino I. Cytological diagnosis of Xp11 translocation renal cell carcinoma: An unusual suspect in bone metastases from unknown primary malignancies. Diagn Cytopathol 2020; 49:11-17. [PMID: 32809251 DOI: 10.1002/dc.24587] [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: 04/30/2020] [Revised: 07/14/2020] [Accepted: 08/03/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Renal cell carcinoma (RCC) constitutes 3% of all cancers, with a higher incidence in patients with age between 60 and 70 years. RCC frequently present as a metastatic tumor at diagnosis, and bones represent one of the most frequent sites. Many cases, mainly in young patients, includes the Xp11 translocation RCC. The cytological diagnosis of Xp11 translocation RCC in adult population it is rarely performed, likely for the morphological overlap with other adult renal cell carcinoma subtypes. METHODS We retrospectively analyze a series of 92 adult patients with metastatic bone tumors, diagnosed on fine-needle aspiration cytology (FNAC) samples, focusing mainly on the cytological, immunophenotypic and molecular features of Xp11 translocation RCC. RESULTS In our series 6 of 92 (6.5%) cases were metastatic RCC (mRCC), among them 2 cases were metastasis from Xp11translocation RCC. Those cases showed a bloody background, with several groups of atypical cells arranged in syncytial groups or in papillary groups composed by atypical cells with abundant cytoplasm, with scattered clear cells. TFE3 was positive on immunocytochemical analysis and specific translocation t(Xp11.23) was detected by FISH analysis. CONCLUSIONS In adult patients with mRCC, it is necessary to consider also Xp11 translocation RCC among the diagnostic hypotheses. FNAC represents a valid tool to investigate bone lesions but cytological features of Xp11 translocation RCC are still poorly described and must necessarily be better defined.
Collapse
Affiliation(s)
- Marco Montella
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Gabriella Aquino
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale," IRCCS, Naples, Nepal
| | - Andrea Ronchi
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Federica Zito Marino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università della Campania "Luigi Vanvitelli", Naples, Italy
| | - Marilena Di Napoli
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Sandro Pignata
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale," IRCCS, Naples, Italy
| | - Immacolata Cozzolino
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università della Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
36
|
Arun G, Aggarwal D, Spector DL. MALAT1 Long Non-Coding RNA: Functional Implications. Noncoding RNA 2020; 6:E22. [PMID: 32503170 PMCID: PMC7344863 DOI: 10.3390/ncrna6020022] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
The mammalian genome is pervasively transcribed and the functional significance of many long non-coding RNA (lncRNA) transcripts are gradually being elucidated. Metastasis Associated Lung Adenocarcinoma Transcript 1 (MALAT1) is one of the most well-studied lncRNAs. MALAT1 is a highly conserved nuclear retained lncRNA that is abundantly expressed in cells and tissues and has been shown to play a role in regulating genes at both the transcriptional and post-transcriptional levels in a context-dependent manner. However, Malat1 has been shown to be dispensable for normal development and viability in mice. Interestingly, accumulating evidence suggests that MALAT1 plays an important role in numerous diseases including cancer. Here, we discuss the current state-of-knowledge in regard to MALAT1 with respect to its function, role in diseases, and the potential therapeutic opportunities for targeting MALAT1 using antisense oligonucleotides and small molecules.
Collapse
Affiliation(s)
- Gayatri Arun
- Envisagenics, 101 Avenue of the Americas, New York, NY 10013, USA;
| | - Disha Aggarwal
- Graduate Program in Genetics, Stony Brook University, Stony Brook, New York, NY 11794, USA;
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724, USA
| | - David L. Spector
- Graduate Program in Genetics, Stony Brook University, Stony Brook, New York, NY 11794, USA;
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724, USA
| |
Collapse
|
37
|
TFEB Modulates p21/WAF1/CIP1 during the DNA Damage Response. Cells 2020; 9:cells9051186. [PMID: 32397616 PMCID: PMC7290768 DOI: 10.3390/cells9051186] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/01/2020] [Accepted: 05/08/2020] [Indexed: 01/01/2023] Open
Abstract
The MiT/TFE family of transcription factors (MITF, TFE3, and TFEB), which control transcriptional programs for autophagy and lysosome biogenesis have emerged as regulators of energy metabolism in cancer. Thus, their activation increases lysosomal catabolic function to sustain cancer cell growth and survival in stress conditions. Here, we found that TFEB depletion dramatically reduces basal expression levels of the cyclin-dependent kinase (CDK) inhibitor p21/WAF1 in various cell types. Conversely, TFEB overexpression increases p21 in a p53-dependent manner. Furthermore, induction of DNA damage using doxorubicin induces TFEB-mediated activation of p21, delays G2/M phase arrest, and promotes cell survival. Pharmacological inhibition of p21, instead, abrogates TFEB-mediated protection during the DNA damage response. Together, our findings uncover a novel and direct role of TFEB in the regulation of p21 expression in both steady-state conditions and during the induction of DNA-damage response (DDR). Our observations might open novel therapeutic strategies to promote cancer cell death by targeting the TFEB-p21 pathway in the presence of genotoxic agents.
Collapse
|
38
|
Abstract
TFEB is overexpressed in TFEB-rearranged renal cell carcinomas as well as in renal tumors with amplifications of TFEB at 6p21.1. As recent literature suggests that renal tumors with 6p21.1 amplification behave more aggressively than those with rearrangements of TFEB, we compared relative TFEB gene expression in these tumors. This study included 37 TFEB-altered tumors: 15 6p21.1-amplified and 22 TFEB-rearranged (including 5 cases from The Cancer Genome Atlas data set). TFEB status was verified using a combination of fluorescent in situ hybridization (n=27) or comprehensive molecular profiling (n=13) and digital droplet polymerase chain reaction was used to quantify TFEB mRNA expression in 6p21.1-amplified (n=9) and TFEB-rearranged renal tumors (n=19). These results were correlated with TFEB immunohistochemistry. TFEB-altered tumors had higher TFEB expression when normalized to B2M (mean: 168.9%, n=28), compared with non-TFEB-altered controls (mean: 7%, n=18, P=0.005). Interestingly, TFEB expression in tumors with rearrangements (mean: 224.7%, n=19) was higher compared with 6p21.1-amplified tumors (mean: 51.2%, n=9; P=0.06). Of note, classic biphasic morphology was only seen in TFEB-rearranged tumors and when present correlated with 6.8-fold higher TFEB expression (P=0.00004). Our results suggest that 6p21.1 amplified renal tumors show increased TFEB gene expression but not as much as t(6;11) renal tumors. These findings correlate with the less consistent/diffuse expression of downstream markers of TFEB activation (cathepsin K, melan A, HMB45) seen in the amplified neoplasms. This suggests that the aggressive biological behavior of 6p21.1 amplified renal tumors might be secondary to other genes at the 6p21.1 locus that are co-amplified, such as VEGFA and CCND3, or other genetic alterations.
Collapse
|
39
|
Bahrami A, Bianconi V, Pirro M, Orafai HM, Sahebkar A. The role of TFEB in tumor cell autophagy: Diagnostic and therapeutic opportunities. Life Sci 2020; 244:117341. [PMID: 31972208 DOI: 10.1016/j.lfs.2020.117341] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/29/2019] [Accepted: 01/18/2020] [Indexed: 12/12/2022]
Abstract
Autophagy is a conserved "self-eating" recycling process which removes aggregated or misfolded proteins, or defective organelles, to maintain cellular hemostasis. In the autophagy-lysosome pathway (ALP), clearance of unwanted debris and materials occurs through the generation of the autophagosome, a complex of double-membrane bounded vesicles that form around cytosolic cargos and catabolize their contents by fusion to lysosomes. In tumors, autophagy has dichotomous functions via preventing tumor initiation but promoting tumor progression. The basic helix-loop-helix leucine zipper transcription factor EB (TFEB) activates the promoters of genes encoding for proteins, which participate in this cellular degradative system by regulating lysosomal biogenesis, lysosomal acidification, lysosomal exocytosis and autophagy. In humans, disturbances of ALP are related to various pathological conditions. Recently, TFEB dysregulation was found to have a crucial pathogenic role in different tumors by modulating tumor cell autophagy. Notably, in renal cell carcinomas, different TFEB gene fusions were reported to promote oncogenic features. In this review, we discuss the role of TFEB in human cancers with a special focus on potential diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Hossein M Orafai
- Department of Pharmaceutics, Faculty of Pharmacy, University of Ahl Al Bayt, Karbala, Iraq; Department of Pharmaceutics, Faculty of Pharmacy, Al-Zahraa University, Karbala, Iraq
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
40
|
Lawrence RE, Fromm SA, Fu Y, Yokom AL, Kim DJ, Thelen AM, Young LN, Lim CY, Samelson AJ, Hurley JH, Zoncu R. Structural mechanism of a Rag GTPase activation checkpoint by the lysosomal folliculin complex. Science 2019; 366:971-977. [PMID: 31672913 DOI: 10.1126/science.aax0364] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/23/2019] [Indexed: 12/16/2022]
Abstract
The tumor suppressor folliculin (FLCN) enables nutrient-dependent activation of the mechanistic target of rapamycin complex 1 (mTORC1) protein kinase via its guanosine triphosphatase (GTPase) activating protein (GAP) activity toward the GTPase RagC. Concomitant with mTORC1 inactivation by starvation, FLCN relocalizes from the cytosol to lysosomes. To determine the lysosomal function of FLCN, we reconstituted the human lysosomal FLCN complex (LFC) containing FLCN, its partner FLCN-interacting protein 2 (FNIP2), and the RagAGDP:RagCGTP GTPases as they exist in the starved state with their lysosomal anchor Ragulator complex and determined its cryo-electron microscopy structure to 3.6 angstroms. The RagC-GAP activity of FLCN was inhibited within the LFC, owing to displacement of a catalytically required arginine in FLCN from the RagC nucleotide. Disassembly of the LFC and release of the RagC-GAP activity of FLCN enabled mTORC1-dependent regulation of the master regulator of lysosomal biogenesis, transcription factor E3, implicating the LFC as a checkpoint in mTORC1 signaling.
Collapse
Affiliation(s)
- Rosalie E Lawrence
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA.,The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Simon A Fromm
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Yangxue Fu
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Adam L Yokom
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Do Jin Kim
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Ashley M Thelen
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA.,The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Lindsey N Young
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Chun-Yan Lim
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA.,The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Avi J Samelson
- The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA.,Institute for Neurodegenerative Diseases, University of California at San Francisco, San Francisco, CA 94158, USA
| | - James H Hurley
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA. .,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94720, USA. .,The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
41
|
Zhang C, Duan Y, Xia M, Dong Y, Chen Y, Zheng L, Chai S, Zhang Q, Wei Z, Liu N, Wang J, Sun C, Tang Z, Cheng X, Wu J, Wang G, Zheng F, Laurence A, Li B, Yang XP. TFEB Mediates Immune Evasion and Resistance to mTOR Inhibition of Renal Cell Carcinoma via Induction of PD-L1. Clin Cancer Res 2019; 25:6827-6838. [PMID: 31383732 DOI: 10.1158/1078-0432.ccr-19-0733] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/11/2019] [Accepted: 08/01/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Cai Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Yaqi Duan
- Department of Pathology, School of Basic Medicine, Tongji Medical College, HUST, Wuhan, China
- Institute of Pathology, Tongji Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Minghui Xia
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Yuting Dong
- Department of Pathology, School of Basic Medicine, Tongji Medical College, HUST, Wuhan, China
- Institute of Pathology, Tongji Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Yufei Chen
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Lu Zheng
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, HUST, Wuhan, China
| | - Shuaishuai Chai
- Department of Urology, Union Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Qian Zhang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, HUST, Wuhan, China
- Institute of Pathology, Tongji Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Zhengping Wei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Na Liu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Jing Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Zhaohui Tang
- Department of Surgery, Tongji Hospital, HUST, Wuhan, China
| | - Xiang Cheng
- Laboratory of Cardiovascular Immunology, Institute of Cardiology, Union Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Guoping Wang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, HUST, Wuhan, China
- Institute of Pathology, Tongji Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Arian Laurence
- Department of Haematology, University College Hospital, London, England
| | - Bing Li
- Department of Urology, Union Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Xiang-Ping Yang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China.
| |
Collapse
|
42
|
Baba M, Furuya M, Motoshima T, Lang M, Funasaki S, Ma W, Sun HW, Hasumi H, Huang Y, Kato I, Kadomatsu T, Satou Y, Morris N, Karim BO, Ileva L, Kalen JD, Wilan Krisna LA, Hasumi Y, Sugiyama A, Kurahashi R, Nishimoto K, Oyama M, Nagashima Y, Kuroda N, Araki K, Eto M, Yao M, Kamba T, Suda T, Oike Y, Schmidt LS, Linehan WM. TFE3 Xp11.2 Translocation Renal Cell Carcinoma Mouse Model Reveals Novel Therapeutic Targets and Identifies GPNMB as a Diagnostic Marker for Human Disease. Mol Cancer Res 2019; 17:1613-1626. [PMID: 31043488 PMCID: PMC6679785 DOI: 10.1158/1541-7786.mcr-18-1235] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/12/2019] [Accepted: 04/26/2019] [Indexed: 12/16/2022]
Abstract
Renal cell carcinoma (RCC) associated with Xp11.2 translocation (TFE3-RCC) has been recently defined as a distinct subset of RCC classified by characteristic morphology and clinical presentation. The Xp11 translocations involve the TFE3 transcription factor and produce chimeric TFE3 proteins retaining the basic helix-loop-helix leucine zipper structure for dimerization and DNA binding suggesting that chimeric TFE3 proteins function as oncogenic transcription factors. Diagnostic biomarkers and effective forms of therapy for advanced cases of TFE3-RCC are as yet unavailable. To facilitate the development of molecular based diagnostic tools and targeted therapies for this aggressive kidney cancer, we generated a translocation RCC mouse model, in which the PRCC-TFE3 transgene is expressed specifically in kidneys leading to the development of RCC with characteristic histology. Expression of the receptor tyrosine kinase Ret was elevated in the kidneys of the TFE3-RCC mice, and treatment with RET inhibitor, vandetanib, significantly suppressed RCC growth. Moreover, we found that Gpnmb (Glycoprotein nonmetastatic B) expression was notably elevated in the TFE3-RCC mouse kidneys as seen in human TFE3-RCC tumors, and confirmed that GPNMB is the direct transcriptional target of TFE3 fusions. While GPNMB IHC staining was positive in 9/9 cases of TFE3-RCC, Cathepsin K, a conventional marker for TFE3-RCC, was positive in only 67% of cases. These data support RET as a potential target and GPNMB as a diagnostic marker for TFE3-RCC. The TFE3-RCC mouse provides a preclinical in vivo model for the development of new biomarkers and targeted therapeutics for patients affected with this aggressive form of RCC. IMPLICATIONS: Key findings from studies with this preclinical mouse model of TFE3-RCC underscore the potential for RET as a therapeutic target for treatment of patients with TFE3-RCC, and suggest that GPNMB may serve as diagnostic biomarker for TFE3 fusion RCC.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Animals
- Apoptosis
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Cycle Proteins/genetics
- Cell Proliferation
- Child
- Chromosomes, Human, X
- Disease Models, Animal
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Middle Aged
- Neoplasm Proteins/genetics
- Oncogene Proteins, Fusion
- Prognosis
- Survival Rate
- Translocation, Genetic
- Tumor Cells, Cultured
- Young Adult
Collapse
Affiliation(s)
- Masaya Baba
- Laboratory of Cancer Metabolism, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan.
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mitsuko Furuya
- Department of Molecular Pathology, Yokohama City University, Yokohama, Japan
| | | | - Martin Lang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Shintaro Funasaki
- Laboratory of Cancer Metabolism, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Wenjuan Ma
- Laboratory of Cancer Metabolism, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Hong-Wei Sun
- Biodata Mining and Discovery Section, NIAMS, NIH, Bethesda, Maryland
| | - Hisashi Hasumi
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Department of Urology, Yokohama City University, Yokohama, Japan
| | - Ying Huang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ikuma Kato
- Department of Molecular Pathology, Yokohama City University, Yokohama, Japan
| | | | - Yorifumi Satou
- Laboratory of Retroviral Genomics and Transcriptomics, International Research Center for Medical Sciences (IRCMS), Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Nicole Morris
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Baktiar O Karim
- Pathology/Histotechnology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Lilia Ileva
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Joseph D Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Luh Ade Wilan Krisna
- Laboratory of Cancer Metabolism, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Yukiko Hasumi
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Aiko Sugiyama
- DSK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Ryoma Kurahashi
- Department of Urology, Kumamoto University, Kumamoto, Japan
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Koshiro Nishimoto
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Masafumi Oyama
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Yoji Nagashima
- Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Naoto Kuroda
- Department of Pathology, Kochi Red Cross Hospital, Kochi, Japan
| | - Kimi Araki
- Division of Developmental Genetics, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Masatoshi Eto
- Department of Urology, Kyushyu University, Fukuoka, Japan
| | - Masahiro Yao
- Department of Urology, Yokohama City University, Yokohama, Japan
| | - Tomomi Kamba
- Department of Urology, Kumamoto University, Kumamoto, Japan
| | - Toshio Suda
- Laboratory of Stem Cell Regulation, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore
| | - Yuichi Oike
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
43
|
Linehan WM, Schmidt LS, Crooks DR, Wei D, Srinivasan R, Lang M, Ricketts CJ. The Metabolic Basis of Kidney Cancer. Cancer Discov 2019; 9:1006-1021. [PMID: 31088840 DOI: 10.1158/2159-8290.cd-18-1354] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/19/2019] [Accepted: 03/22/2019] [Indexed: 01/27/2023]
Abstract
Kidney cancer is not a single disease but represents several distinct types of cancer that have defining histologies and genetic alterations and that follow different clinical courses and have different responses to therapy. Mutation of genes associated with kidney cancer, such as VHL, FLCN, TFE3, FH, or SDHB, dysregulates the tumor's responses to changes in oxygen, iron, nutrient, or energy levels. The identification of these varying genetic bases of kidney cancer has increased our understanding of the biology of this cancer, allowing the development of targeted therapies and the appreciation that it is a cancer driven by metabolic alterations. SIGNIFICANCE: Kidney cancer is a complex disease composed of different types of cancer that present with different histologies, clinical courses, genetic changes, and responses to therapy. This review describes the known genetic changes within kidney cancer, how they alter tumor metabolism, and how these metabolic changes can be therapeutically targeted.
Collapse
Affiliation(s)
- W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.,Basic Science Program, Frederick Laboratory for Cancer Research, Frederick, Maryland
| | - Daniel R Crooks
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Darmood Wei
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Martin Lang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Christopher J Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
44
|
Xie L, Zhang Y, Wu CL. Microphthalmia family of transcription factors associated renal cell carcinoma. Asian J Urol 2019; 6:312-320. [PMID: 31768316 PMCID: PMC6872788 DOI: 10.1016/j.ajur.2019.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/25/2018] [Accepted: 11/06/2018] [Indexed: 12/11/2022] Open
Abstract
The microphthalmia (MiT) subfamily of transcription factors includes TFE3, TFEB, TFEC, and MITF. In the 2016 World Health Organization classification, MiT family translocation renal cell carcinoma (tRCC) including Xp11 tRCC and t(6;11) RCC, was newly defined as an RCC subtype. Xp11 and t(6;11) RCC are characterized by the rearrangement of the MiT transcription factors TFE3 and TFEB, respectively. Recent studies identified the fusion partner-dependent clinicopathological and immunohistochemical features in TFE3-rearranged RCC. Furthermore, RCC with TFEB amplification, melanotic MiT family translocation neoplasms, was identified may as a unique subtype of MiT family associated renal neoplasms, along with MITF associated RCC. In this review, we will collect available literature of these newly-described RCCs, analyze their clinicopathological and immunohistochemical features, and summarize their molecular and genetic evidences. We expect this review would be beneficial for the understanding of these rare subtypes of RCCs, and eventually promote clinical management strategies.
Collapse
Affiliation(s)
- Ling Xie
- Department of Pathology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Department of Pathology and Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yifen Zhang
- Department of Pathology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chin-Lee Wu
- Department of Pathology and Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
NEAT1-TFE3 and KAT6A-TFE3 renal cell carcinomas, new members of MiT family translocation renal cell carcinoma. Mod Pathol 2019; 32:710-716. [PMID: 30622287 PMCID: PMC6486435 DOI: 10.1038/s41379-018-0191-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 12/31/2022]
Abstract
Microphthalmia-associated transcription factor (MiT) family translocation renal cell carcinoma harbors variable gene fusions involving either TFE3 or TFEB genes. Multiple 5' fusion partners for TFE3 have been reported, including ASPSCR1, CLTC, DVL2, LUC7L3, KHSRP, PRCC, PARP14, NONO, SFPQ1, MED15, and RBM10. Each of these fusion genes activates TFE3 transcription which can be detected by immunostaining. Using targeted RNA-sequencing, TFE3 fusion gene partners were identified in 5 cases of TFE3 immunohistochemistry positive translocation renal cell carcinoma. Three cases demonstrated known fusions: ASPSCR1-TFE3, MED15-TFE3 and RBM10-TFE3. However, two cases showed unreported NEAT1-TFE3 and KAT6A-TFE3 fusion transcripts. The NEAT1-TFE3 RCC arose in a 59-year-old male; which demonstrated overlapping morphological features seen in NEAT2(MALAT1)-TFEB t(6;11) renal cell carcinoma, including biphasic alveolar/nested tumor cells with eosinophilic cytoplasm. The KAT6A-TFE3 renal cell carcinoma demonstrated typical morphological features of TFE3/Xp11 renal cell carcinoma including papillae, eosinophilic cytoplasm with focal clearing and abundant psammoma bodies. KAT6A gene fusion was reported in some cases of acute myeloid leukemia, which has not been previously reported in solid tumors. This report highlights the genetic complexity of TFE3 translocation renal cell carcinoma; and RNA-sequencing is a powerful approach for elucidating the underlying genetic alterations.
Collapse
|
46
|
Tuna M, Amos CI, Mills GB. Molecular mechanisms and pathobiology of oncogenic fusion transcripts in epithelial tumors. Oncotarget 2019; 10:2095-2111. [PMID: 31007851 PMCID: PMC6459343 DOI: 10.18632/oncotarget.26777] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 02/22/2019] [Indexed: 02/07/2023] Open
Abstract
Recurrent fusion transcripts, which are one of the characteristic hallmarks of cancer, arise either from chromosomal rearrangements or from transcriptional errors in splicing. DNA rearrangements include intrachromosomal or interchromosomal translocation, tandem duplication, deletion, inversion, or result from chromothripsis, which causes complex rearrangements. In addition, fusion proteins can be created through transcriptional read-through. Fusion genes can be transcribed to fusion transcripts and translated to chimeric proteins, with many having demonstrated transforming activities through multiple mechanisms in cells. Fusion proteins represent novel therapeutic targets and diagnostic biomarkers of diagnosis, disease status, or progression. This review focuses on the mechanisms underlying the formation of oncogenic fusion genes and transcripts and their impact on the pathobiology of epithelial tumors.
Collapse
Affiliation(s)
- Musaffe Tuna
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Christopher I. Amos
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Gordon B. Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cell, Developmental and Cancer Biology, School of Medicine, Oregon Health Science University, Portland, OR, USA
- Precision Oncology, Knight Cancer Institute, Portland, OR, USA
| |
Collapse
|
47
|
Perera RM, Di Malta C, Ballabio A. MiT/TFE Family of Transcription Factors, Lysosomes, and Cancer. ANNUAL REVIEW OF CANCER BIOLOGY 2019; 3:203-222. [PMID: 31650096 PMCID: PMC6812561 DOI: 10.1146/annurev-cancerbio-030518-055835] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer cells have an increased demand for energy sources to support accelerated rates of growth. When nutrients become limiting, cancer cells may switch to nonconventional energy sources that are mobilized through nutrient scavenging pathways involving autophagy and the lysosome. Thus, several cancers are highly reliant on constitutive activation of these pathways to degrade and recycle cellular materials. Here, we focus on the MiT/TFE family of transcription factors, which control transcriptional programs for autophagy and lysosome biogenesis and have emerged as regulators of energy metabolism in cancer. These new findings complement earlier reports that chromosomal translocations and amplifications involving the MiT/TFE genes contribute to the etiology and pathophysiology of renal cell carcinoma, melanoma, and sarcoma, suggesting pleiotropic roles for these factors in a wider array of cancers. Understanding the interplay between the oncogenic and stress-adaptive roles of MiT/TFE factors could shed light on fundamental mechanisms of cellular homeostasis and point to new strategies for cancer treatment.
Collapse
Affiliation(s)
- Rushika M Perera
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143, USA
| | - Chiara Di Malta
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, 80138Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, 80138Naples, Italy
- Department of Molecular and Human Genetics and Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
48
|
Wyvekens N, Rechsteiner M, Fritz C, Wagner U, Tchinda J, Wenzel C, Kuithan F, Horn LC, Moch H. Histological and molecular characterization of TFEB-rearranged renal cell carcinomas. Virchows Arch 2019; 474:625-631. [PMID: 30706129 DOI: 10.1007/s00428-019-02526-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/27/2018] [Accepted: 01/14/2019] [Indexed: 01/07/2023]
Abstract
The 2016 WHO Classification of Tumors of the Urinary System recognizes microphthalmia transcription factor (MiT) family translocation carcinomas as a separate entity among renal cell carcinomas. TFE3 and transcription factor EB (TFEB) are members of the MiT family for which chromosomal rearrangements have been associated with renal cell carcinoma formation. TFEB translocation renal cell carcinoma is a rare tumor harboring a t(6;11)(p21;q12) translocation. Recently, renal cell carcinomas with TFEB amplification have been identified. TFEB amplified renal cell carcinomas have to be distinguished from TFEB-translocated renal cancer, because they may demonstrate a more aggressive behavior. Herein, we present a TFEB-translocated and a TFEB-amplified carcinoma cases and describe their distinct histological, immunohistochemical, and molecular characteristics. In addition, we review conventional morphology, immunophenotype, genetic background, and clinical outcome of TFEB-rearranged RCCs in the literature, with a special emphasis on important differential diagnoses and the diagnostic approach.
Collapse
Affiliation(s)
- Nicolas Wyvekens
- Department of Pathology and Molecular Pathology, University Hospital and University Zurich, Schmelzbergstrasse 12, 8091, Zurich, Switzerland
| | - Markus Rechsteiner
- Department of Pathology and Molecular Pathology, University Hospital and University Zurich, Schmelzbergstrasse 12, 8091, Zurich, Switzerland
| | - Christine Fritz
- Department of Pathology and Molecular Pathology, University Hospital and University Zurich, Schmelzbergstrasse 12, 8091, Zurich, Switzerland
| | - Ulrich Wagner
- Department of Pathology and Molecular Pathology, University Hospital and University Zurich, Schmelzbergstrasse 12, 8091, Zurich, Switzerland
| | - Joëlle Tchinda
- Department of Oncology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Carina Wenzel
- Department of Pathology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Friederike Kuithan
- Department of Pathology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | | | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital and University Zurich, Schmelzbergstrasse 12, 8091, Zurich, Switzerland.
| |
Collapse
|
49
|
Abstract
PURPOSE The research of long non-coding RNAs (lncRNAs) has become a new passion with the discovery of abundant new lncRNAs and extensive investigation of their roles in various diseases, especially in cancers. Metastasis associated in lung adenocarcinoma transcript 1 (MALAT1) emerges as a hotspot, which has been reported to be involved in dysregulation of cell signaling and closely correlated with cancer development, progression, and response to therapy. This review is a brief update of the current knowledge related to the role of MALAT1 in cancer-associated molecular pathways and pathophysiology and possible determinants for MALAT1 to function as a biomarker, aiming to stimulate the basic investigation of lncRNA MALAT1 as well as its translation to clinical applications. METHODS We have selected vast literature from electronic databases including studies associated with its clinical significance and the pivotal functions in cancer processes such as cell proliferation, apoptosis, metastasis, immunity, angiogenesis, and drug resistance. RESULTS Studies have shown that aberrant expression of MALAT1 is related to cancer pathophysiology with the potential to be translated clinically and MALAT1 can regulate cancer processes by interacting with molecules, such as proteins, RNAs and DNAs, and further altering different signal pathways. CONCLUSION MALAT1 lncRNA promises to be a potential biomarker for cancer diagnosis as well as prognosis. Additionally, it might be a therapeutic target for human cancers.
Collapse
Affiliation(s)
- Zhi-Xing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China,
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410078, People's Republic of China,
| | - Qiong-Ni Zhu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China,
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410078, People's Republic of China,
| | - Hai-Bo Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China,
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410078, People's Republic of China,
| | - Yang Hu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China,
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410078, People's Republic of China,
| | - Guo Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China,
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410078, People's Republic of China,
| | - Yuan-Shan Zhu
- Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA,
| |
Collapse
|
50
|
Kulkarni P, Dasgupta P, Bhat NS, Shahryari V, Shiina M, Hashimoto Y, Majid S, Deng G, Saini S, Tabatabai ZL, Yamamura S, Tanaka Y, Dahiya R. Elevated miR-182-5p Associates with Renal Cancer Cell Mitotic Arrest through Diminished MALAT-1 Expression. Mol Cancer Res 2018; 16:1750-1760. [PMID: 30037856 DOI: 10.1158/1541-7786.mcr-17-0762] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/03/2018] [Accepted: 07/03/2018] [Indexed: 12/23/2022]
Abstract
The molecular heterogeneity of clear cell renal carcinoma (ccRCC) makes prediction of disease progression and therapeutic response difficult. Thus, this report investigates the functional significance, mechanisms of action, and clinical utility of miR-182-5p and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1/NEAT2), a long noncoding RNA (lncRNA), in the regulation of kidney cancer using human kidney cancer tissues as well as in vitro and in vivo model systems. Profiling of miR-182-5p and MALAT-1 in human renal cancer cells and clinical specimens was done by quantitative real-time PCR (qPCR). The biological significance was determined by series of in vitro and in vivo experiments. The interaction between miR-182-5p and MALAT-1 was investigated using luciferase reporter assays. In addition, the effects of miR-182-5p overexpression and MALAT-1 downregulation on cell-cycle progression were assessed in ccRCC cells. The data indicate that miR-182-5p is downregulated in ccRCC; the mechanism being CpG hypermethylation as observed from 5-Aza CdR treatment that decreased promoter methylation and expression of key methylation regulatory genes like DNMT1, DNMT3a, and DNMT3b Overexpression of miR-182-5p-inhibited cell proliferation, colony formation, apoptosis, and led to G2-M-phase cell-cycle arrest by directly targeting MALAT-1 Downregulation of MALAT-1 led to upregulation of p53, downregulation of CDC20, AURKA, drivers of the cell-cycle mitotic phase. Transient knockdown of MALAT-1 mimicked the effects of miR-182-5p overexpression. Finally, overexpression of miR-182-5p decreased tumor growth in mice, compared with controls; thus, demonstrating its antitumor effect in vivo Implications: This is the first study that offers new insight into role of miR-182-5p/MALAT-1 interaction on inhibition of ccRCC progression. Mol Cancer Res; 16(11); 1750-60. ©2018 AACR.
Collapse
Affiliation(s)
- Priyanka Kulkarni
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Pritha Dasgupta
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Nadeem S Bhat
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, Florida
| | - Varahram Shahryari
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Marisa Shiina
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Yutaka Hashimoto
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Shahana Majid
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Guoren Deng
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Sharanjot Saini
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Z Laura Tabatabai
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Soichiro Yamamura
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Yuichiro Tanaka
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California
- University of California San Francisco, San Francisco, California
| | - Rajvir Dahiya
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California.
- University of California San Francisco, San Francisco, California
| |
Collapse
|