75 results on '"Shukuan Li"'
Search Results
2. Acetylation at lysine 27 on maternal H3.3 regulates minor zygotic genome activation
- Author
-
Jiaming Zhang, Xuanwen Li, Qi Zhao, Jingzhang Ji, Hongdi Cui, Weibo Hou, Xinyu Wang, Entong Song, Songling Xiao, Shukuan Ling, Shaorong Gao, Xiaoyu Liu, Duancheng Wen, and Qingran Kong
- Subjects
CP: Molecular biology ,CP: Developmental biology ,Biology (General) ,QH301-705.5 - Abstract
Summary: Zygotic genome activation (ZGA) initiates transcription in early embryogenesis and requires extensive chromatin remodeling, including rapid incorporation of the histone variant H3.3. The distinct sources of H3.3 from paternal and maternal alleles (paH3.3 and maH3.3) complicate tracking their individual contributions. Here, using an H3.3B-hemagglutinin (HA)-tagged mouse model, we profile the temporal dynamics of paH3.3 and maH3.3, revealing a unique pattern of maH3.3 enrichment at the promoter regions from zygotes to 2-cell embryos, highlighting the crucial role of maternally stored H3.3 mRNAs and proteins (mH3.3) in pre-implantation development. Knockdown of mH3.3 compromises cleavage and minor ZGA. Mechanistically, mH3.3 facilitates minor ZGA through H3.3S31ph-dependent H3K27ac deposition. Profiling of H3.3 landscape in parthenogenetic (PG) and androgenetic (AG) embryos highlights the role of mH3.3 in remodeling the paternal genome by establishing H3K27ac. These findings demonstrate that mH3.3-mediated parental chromatin reprogramming is essential for orchestrating minor ZGA.
- Published
- 2025
- Full Text
- View/download PDF
3. Ckip-1 3’UTR alleviates prolonged sleep deprivation induced cardiac dysfunction by activating CaMKK2/AMPK/cTNI pathway
- Author
-
Beilei Dong, Rui Xue, Jianwei Li, Shukuan Ling, Wenjuan Xing, Zizhong Liu, Xinxin Yuan, Junjie Pan, Ruikai Du, Xinming Shen, Jingwen Zhang, Youzhi Zhang, Yingxian Li, and Guohui Zhong
- Subjects
Sleep deprivation ,Cardiac function ,Ckip-1 3'UTR ,CaMKK2/AMPK/cTNI pathway ,Medicine - Abstract
Abstract Sleep deprivation (SD) has emerged as a critical concern impacting human health, leading to significant damage to the cardiovascular system. However, the underlying mechanisms are still unclear, and the development of targeted drugs is lagging. Here, we used mice to explore the effects of prolonged SD on cardiac structure and function. Echocardiography analysis revealed that cardiac function was significantly decreased in mice after five weeks of SD. Real-time quantitative PCR (RT-q-PCR) and Masson staining analysis showed that cardiac remodeling marker gene Anp (atrial natriuretic peptide) and fibrosis were increased, Elisa assay of serum showed that the levels of creatine kinase (CK), creatine kinase-MB (CK-MB), ANP, brain natriuretic peptide (BNP) and cardiac troponin T (cTn-T) were increased after SD, suggesting that cardiac remodeling and injury occurred. Transcript sequencing analysis indicated that genes involved in the regulation of calcium signaling pathway, dilated cardiomyopathy, and cardiac muscle contraction were changed after SD. Accordingly, Western blotting analysis demonstrated that the cardiac-contraction associated CaMKK2/AMPK/cTNI pathway was inhibited. Since our preliminary research has confirmed the vital role of Casein Kinase-2 -Interacting Protein-1 (CKIP-1, also known as PLEKHO1) in cardiac remodeling regulation. Here, we found the levels of the 3’ untranslated region of Ckip-1 (Ckip-1 3’UTR) decreased, while the coding sequence of Ckip-1 (Ckip-1 CDS) remained unchanged after SD. Significantly, adenovirus-mediated overexpression of Ckip-1 3’UTR alleviated SD-induced cardiac dysfunction and remodeling by activating CaMKK2/AMPK/cTNI pathway, which proposed the therapeutic potential of Ckip-1 3’UTR in treating SD-induced heart disease.
- Published
- 2024
- Full Text
- View/download PDF
4. Lightweight Deep Learning Model, ConvNeXt-U: An Improved U-Net Network for Extracting Cropland in Complex Landscapes from Gaofen-2 Images
- Author
-
Shukuan Liu, Shi Cao, Xia Lu, Jiqing Peng, Lina Ping, Xiang Fan, Feiyu Teng, and Xiangnan Liu
- Subjects
fragmented cropland extraction ,ConvNeXt-U ,lightweight model ,GF-2 ,remote sensing ,Chemical technology ,TP1-1185 - Abstract
Extracting fragmented cropland is essential for effective cropland management and sustainable agricultural development. However, extracting fragmented cropland presents significant challenges due to its irregular and blurred boundaries, as well as the diversity in crop types and distribution. Deep learning methods are widely used for land cover classification. This paper proposes ConvNeXt-U, a lightweight deep learning network that efficiently extracts fragmented cropland while reducing computational requirements and saving costs. ConvNeXt-U retains the U-shaped structure of U-Net but replaces the encoder with a simplified ConvNeXt architecture. The decoder remains unchanged from U-Net, and the lightweight CBAM (Convolutional Block Attention Module) is integrated. This module adaptively adjusts the channel and spatial dimensions of feature maps, emphasizing key features and suppressing redundant information, which enhances the capture of edge features and improves extraction accuracy. The case study area is Hengyang County, Hunan Province, China, using GF-2 remote sensing imagery. The results show that ConvNeXt-U outperforms existing methods, such as Swin Transformer (Acc = 85.1%, IoU = 79.1%), MobileNetV3 (Acc = 83.4%, IoU = 77.6%), VGG16 (Acc = 80.5%, IoU = 74.6%), and ResUnet (Acc = 81.8%, IoU = 76.1%), achieving an IoU of 79.5% and Acc of 85.2%. Under the same conditions, ConvNeXt-U has a faster inference speed of 37 images/s, compared to 28 images/s for Swin Transformer, 35 images/s for MobileNetV3, and 0.43 and 0.44 images/s for VGG16 and ResUnet, respectively. Moreover, ConvNeXt-U outperforms other methods in processing the boundaries of fragmented cropland, producing clearer and more complete boundaries. The results indicate that the ConvNeXt and CBAM modules significantly enhance the accuracy of fragmented cropland extraction. ConvNeXt-U is also an effective method for extracting fragmented cropland from remote sensing imagery.
- Published
- 2025
- Full Text
- View/download PDF
5. Oral recombinant methioninase (o-rMETase) is superior to injectable rMETase and overcomes acquired gemcitabine resistance in pancreatic cancer
- Author
-
Kentaro Miyake, Kei Kawaguchi, Zhiying Zhang, Shree Ram Singh, Takashi Higuchi, Michiaki Unno, Masuyo Miyake, Yuying Tan, Hiromichi Oshiro, Sahar Razmjooei, Shukuan Li, Sintawat Wangsiricharoen, Kentaro Igarashi, Robert M. Hoffman, Michael Bouvet, Tasuku Kiyuna, and Qinghong Han
- Subjects
0301 basic medicine ,Antimetabolites, Antineoplastic ,Cancer Research ,Administration, Oral ,Mice, Nude ,Apoptosis ,Deoxycytidine ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Nude mouse ,Pancreatic cancer ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Cell Proliferation ,biology ,business.industry ,Cell growth ,Melanoma ,Cancer ,medicine.disease ,biology.organism_classification ,Xenograft Model Antitumor Assays ,Gemcitabine ,Recombinant Proteins ,Pancreatic Neoplasms ,Carbon-Sulfur Lyases ,030104 developmental biology ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,business ,Injections, Intraperitoneal ,medicine.drug - Abstract
Recombinant methioninase (rMETase) was previously administered as an injectable drug to target methionine dependence of cancer. Recently, we observed that rMETase could be administered orally (o-rMETase) in a patient-derived orthotopic xenograft (PDOX) mouse model of melanoma. Here, we determined the efficacy of o-rMETase on a pancreatic cancer PDOX model. Forty pancreatic cancer PDOX mouse models were randomized into four groups of 10 mice each. o-rMETase was significantly more effective than i.p.-rMETase, but the combination of both was significantly more effective than either alone. Acquired gemcitabine resistance is a major factor in the recalcitrance of pancreatic cancer. We tested a human pancreatic cancer cell line, which has acquired >100-fold GEM-resistance (PK-9R) than its parental cell line PK-9. In contrast to GEM, both cell lines were very sensitive to rMETase. In orthotopic nude mouse models of PK-9 and PK-9R, GEM inhibited tumor growth in PK-9 but not PK-9R. In contrast, o-rMETase could inhibit both tumors. The combination of GEM + o-rMETase could regress the PK-9 tumor and inhibit PK-9R tumor growth. The present study shows that o-rMETase is effective and overcomes acquired GEM resistance in pancreatic cancer and demonstrates the clinical potential of this strategy.
- Published
- 2018
- Full Text
- View/download PDF
6. Recombinant methioninase combined with doxorubicin (DOX) regresses a DOX-resistant synovial sarcoma in a patient-derived orthotopic xenograft (PDOX) mouse model
- Author
-
Scott D. Nelson, Tara A. Russell, Qinghong Han, Takashi Higuchi, Hiroyuki Tsuchiya, Shukuan Li, Sarah M. Dry, Kei Kawaguchi, Robert M. Hoffman, Shinji Miwa, Fritz C. Eilber, Mark A. Eckardt, Kentaro Igarashi, Emily Gainor, Kentaro Miyake, Masuyo Miyake, Yunfeng Li, Hiroaki Kimura, Yuying Tan, Katsuhiro Hayashi, Norio Yamamoto, Arun S. Singh, Hiromichi Oshiro, and Tasuku Kiyuna
- Subjects
0301 basic medicine ,Oncology and Carcinogenesis ,Pharmacology ,synovial sarcoma ,Body weight ,doxorubicin ,law.invention ,PDOX ,03 medical and health sciences ,Rare Diseases ,0302 clinical medicine ,Untreated control ,law ,polycyclic compounds ,medicine ,Doxorubicin ,patient-derived orthotopic xenograft ,Cancer ,business.industry ,Soft tissue sarcoma ,recombinant methioninase ,medicine.disease ,Synovial sarcoma ,Orphan Drug ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Recombinant DNA ,Right biceps femoris muscle ,business ,After treatment ,Research Paper ,medicine.drug - Abstract
Synovial sarcoma (SS) is a recalcitrant subgroup of soft tissue sarcoma (STS). A tumor from a patient with high grade SS from a lower extremity was grown orthotopically in the right biceps femoris muscle of nude mice to establish a patient-derived orthotopic xenograft (PDOX) mouse model. The PDOX mice were randomized into the following groups when tumor volume reached approximately 100 mm3: G1, control without treatment; G2, doxorubicin (DOX) (3 mg/kg, intraperitoneal [i.p.] injection, weekly, for 2 weeks; G3, rMETase (100 unit/mouse, i.p., daily, for 2 weeks); G4 DOX (3mg/kg), i.p. weekly, for 2 weeks) combined with rMETase (100 unit/mouse, i.p., daily, for 2 weeks). On day 14 after treatment initiation, all therapies significantly inhibited tumor growth compared to untreated control, except DOX: (DOX: p = 0.48; rMETase: p < 0.005; DOX combined with rMETase < 0.0001). DOX combined with rMETase was significantly more effective than both DOX alone (p < 0.001) and rMETase alone (p < 0.05). The relative body weight on day 14 compared with day 0 did not significantly differ between any treatment group or untreated control. The results indicate that r-METase can overcome DOX-resistance in this recalcitrant disease.
- Published
- 2018
- Full Text
- View/download PDF
7. Recombinant methioninase in combination with doxorubicin (DOX) overcomes first-line DOX resistance in a patient-derived orthotopic xenograft nude-mouse model of undifferentiated spindle-cell sarcoma
- Author
-
Shree Ram Singh, Shukuan Li, Hiroaki Kimura, Sarah M. Dry, Mark A. Eckardt, Hiroyuki Tsuchiya, Fritz C. Eilber, Arun S. Singh, Scott D. Nelson, Kentaro Igarashi, Kei Kawaguchi, Tara A. Russell, Shinji Miwa, Norio Yamamoto, Takashi Murakami, Robert M. Hoffman, Yuying Tan, Tasuku Kiyuna, Yunfeng Li, Masuyo Miyake, Qinghong Han, Kentaro Miyake, and Katsuhiro Hayashi
- Subjects
0301 basic medicine ,Cancer Research ,First line ,Mice, Nude ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Nude mouse ,law ,Antineoplastic Combined Chemotherapy Protocols ,polycyclic compounds ,Animals ,Humans ,Medicine ,Doxorubicin ,biology ,Tumor size ,business.industry ,Body Weight ,Cancer ,Sarcoma ,medicine.disease ,biology.organism_classification ,Xenograft Model Antitumor Assays ,Recombinant Proteins ,Tumor Burden ,Carbon-Sulfur Lyases ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Recombinant DNA ,Spindle cell sarcoma ,business ,Undifferentiated spindle cell sarcoma ,medicine.drug - Abstract
We have previously established a patient-derived orthotopic xenograft (PDOX) model of undifferentiated spindle cell sarcoma (USCS). Recombinant methioninase (rMETase) has previously demonstrated efficacy in PDOX mouse models of human cancers. In the present study, we determined if rMETase in combination with doxorubicin (DOX) can overcome first-line DOX resistance in a PDOX models of USCS. The USCS PDOX mouse models were randomized into the following groups when tumor volume reached 100 mm3: G1, control without treatment; G2, doxorubicin (DOX) (3 mg/kg, intraperitoneal [i.p.] injection, weekly, for 2 weeks); G3, rMETase (100 units/mouse, i.p., daily, for 2 weeks); G4, DOX (3 mg/kg, i.p., weekly, for 2 weeks) combined with rMETase (100 units/mouse, i.p., daily, for 2 weeks). Tumor size and body weight were measured twice a week. On day 14 after initiation, the USCS PDOX tumor sizes were (G1): 360 ± 85 mm3; DOX (G2): 355 ± 111 mm3, p = .927; rMETase (G3): 182 ± 57 mm3, p = .0003; DOX + rMETase (G4): 134 ± 29 mm3, p = .00001. These results indicate that rMETase can overcome USCS resistance to DOX, which is first line therapy for this disease. The body weight of treated mice was not significantly different in any group. The present results demonstrate the power of the PDOX model to identify effective therapy for recalcitrant cancer and the potential of rMETase to overcome DOX resistance.
- Published
- 2018
- Full Text
- View/download PDF
8. Combining Tumor-Selective Bacterial Therapy with Salmonella typhimurium A1-R and Cancer Metabolism Targeting with Oral Recombinant Methioninase Regressed an Ewing’s Sarcoma in a Patient-Derived Orthotopic Xenograft Model
- Author
-
Takashi Chishima, Yuying Tan, Ryusei Matsuyama, Kentaro Miyake, Kei Kawaguchi, Scott D. Nelson, Zhiying Zhang, Shree Ram Singh, Robert M. Hoffman, Tasuku Kiyuna, Yukihiko Hiroshima, Michael Bouvet, Sahar Razmjooei, Shukuan Li, Yunfeng Li, Maryam Barangi, Ming Zhao, Fritz C. Eilber, Takashi Murakami, Hiromichi Oshiro, Takashi Higuchi, Sintawat Wangsiricharoen, Arun S. Singh, Itaru Endo, and Qinghong Han
- Subjects
0301 basic medicine ,Salmonella ,medicine.disease_cause ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Oral administration ,law ,Drug Discovery ,Medicine ,Pharmacology (medical) ,Tumor growth ,Doxorubicin ,Pharmacology ,business.industry ,Ewing's sarcoma ,General Medicine ,medicine.disease ,030104 developmental biology ,Infectious Diseases ,Oncology ,030220 oncology & carcinogenesis ,Cancer metabolism ,Cancer research ,Recombinant DNA ,Sarcoma ,business ,medicine.drug - Abstract
Background: Ewing’s sarcoma (ES) is a recalcitrant disease in need of transformative therapeutics. Objectives: The aim of this study was to investigate the efficacy of tumor-selective Salmonella typhimurium A1-R combined with tumor metabolism targeting with oral administration of recombinant methioninase (o-rMETase), on an ES patient-derived orthotopic xenograft (PDOX) model. Methods: The ES PDOX models were previously established in the right chest wall. The ES PDOX models were randomized into 5 groups when the tumor volume reached 80 mm3: G1: untreated control; G2: doxorubicin; G3: S. typhimurium A1-R; G4: o-rMETase; G5: S. typhimurium A1-R combined with o-rMETase. All mice were sacrificed on day 15. Body weight and tumor volume were assessed twice a week. Results: S. typhimurium A1-R and o-rMETase respectively suppressed tumor growth as monotherapies (p = 0.050 and p = 0.032). S. typhimurium A1-R combined with o-rMETase regressed tumor growth significantly compared to untreated group on day 15 (p < 0.032). S. typhimurium A1-R combined with o-rMETase group was significantly more effective than S. typhimurium A1-R or o-rMETase monotherapy (p = 0.032, p = 0.032). Conclusions: The present results suggest that the combination of S. typhimurium A1-R and o-rMETase has promise to be a transformative therapy for ES.
- Published
- 2018
- Full Text
- View/download PDF
9. Recombinant methioninase (rMETase) is an effective therapeutic for BRAF-V600E-negative as well as -positive melanoma in patient-derived orthotopic xenograft (PDOX) mouse models
- Author
-
Kentaro Igarashi, Fritz C. Eilber, Yunfeng Li, Tara A. Russell, Kei Kawaguchi, Bartosz Chmielowski, Sarah M. Dry, Michiaki Unno, Kentaro Miyake, Masuyo Miyake, Tasuku Kiyuna, Scott D. Nelson, Qinghong Han, Takashi Murakami, Robert M. Hoffman, Yuying Tan, and Shukuan Li
- Subjects
0301 basic medicine ,Combination therapy ,medicine.medical_treatment ,Oncology and Carcinogenesis ,Mutant ,law.invention ,PDOX ,03 medical and health sciences ,0302 clinical medicine ,law ,melanoma ,BRAF-V600E mutation ,Medicine ,Vemurafenib ,neoplasms ,methionine dependence ,Chemotherapy ,Temozolomide ,business.industry ,Melanoma ,Cancer ,recombinant methioninase ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Recombinant DNA ,Cancer research ,business ,Research Paper ,medicine.drug - Abstract
Melanoma is a recalcitrant disease. Melanoma patients with the BRAF-V600E mutation have been treated with the drug vemurafenib (VEM) which targets this mutation. However, we previously showed that VEM is not very effective against a BRAF-V600E melanoma mutant in a patient-derived orthotopic xenograft (PDOX) model. In contrast, we demonstrated that recombinant methioninase (rMETase) which targets the general metabolic defect in cancer of methionine dependence, was effective against the BRAF-V600E mutant melanoma PDOX model. In the present study, we demonstrate that rMETase is effective against a BRAF-V600E-negative melanoma PDOX which we established. Forty BRAF-V600E-negative melanoma PDOX mouse models were randomized into four groups of 10 mice each: untreated control (n = 10); temozolomide (TEM) (25 mg/kg, p.o., 14 consecutive days, n = 10); rMETase (100 units, i.p., 14 consecutive days, n = 10); TEM + rMETase (TEM: 25 mg/kg, p.o., rMETase: 100 units, i.p., 14 consecutive days, n = 10). All treatments inhibited tumor growth compared to untreated control (TEM: p = 0.0003, rMETase: p = 0.0006, TEM/rMETase: p = 0.0002) on day 14 after initiation. Combination therapy of TEM and rMETase was significantly more effective than either mono-therapy (TEM: p = 0.0113, rMETase: p = 0.0173). The present study shows that TEM combined with rMETase is effective for BRAF-V600E-negative melanoma PDOX similar to the BRAF-V600E-positive mutation melanoma. These results suggest rMETase in combination with first-line chemotherapy can be highly effective in both BRAF-V600E-negative as well as BRAF-V600E-positive melanoma and has clinical potential for this recalcitrant disease.
- Published
- 2017
- Full Text
- View/download PDF
10. Combination treatment with recombinant methioninase enables temozolomide to arrest a BRAF V600E melanoma in a patient-derived orthotopic xenograft (PDOX) mouse model
- Author
-
Fritz C. Eilber, Sarah M. Dry, Qinghong Han, Takashi Murakami, Kentaro Miyake, Scott D. Nelson, Michiaki Unno, Shukuan Li, Robert M. Hoffman, Yunfeng Li, Bartosz Chmielowski, Kentaro Igarashi, Kei Kawaguchi, Tara A. Russell, Yuying Tan, and Tasuku Kiyuna
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Combination therapy ,Oncology and Carcinogenesis ,temozolomide ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Nude mouse ,Surgical oncology ,law ,Internal medicine ,melanoma ,medicine ,metabolic targeting ,methionine dependence ,Temozolomide ,Methionine ,biology ,business.industry ,Melanoma ,Cancer ,recombinant methioninase ,biology.organism_classification ,medicine.disease ,3. Good health ,Surgery ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Recombinant DNA ,business ,Research Paper ,medicine.drug - Abstract
// Kei Kawaguchi 1, 2, 3 , Kentaro Igarashi 1, 2 , Shukuan Li 1 , Qinghong Han 1 , Yuying Tan 1 , Tasuku Kiyuna 1, 2 , Kentaro Miyake 1 , 2 , Takashi Murakami 1, 2 , Bartosz Chmielowski 4 , Scott D. Nelson 5 , Tara A. Russell 6 , Sarah M. Dry 5 , Yunfeng Li 5 , Michiaki Unno 3 , Fritz C. Eilber 6 and Robert M. Hoffman 1, 2 1 AntiCancer, Inc., San Diego, CA, USA 2 Department of Surgery, University of California, San Diego, CA, USA 3 Department of Surgery, Graduate School of Medicine, Tohoku University, Sendai, Japan 4 Division of Hematology-Oncology, University of California, Los Angeles, CA, USA 5 Department of Pathology, University of California, Los Angeles, CA, USA 6 Division of Surgical Oncology, University of California, Los Angeles, CA, USA Correspondence to: Robert M. Hoffman, email: all@anticancer.com Fritz C. Eilber, email: fceilber@mednet.ucla.edu Keywords: recombinant methioninase, methionine dependence, metabolic targeting, temozolomide, melanoma Received: June 06, 2017 Accepted: July 06, 2017 Published: August 12, 2017 ABSTRACT An excessive requirement for methionine termed methionine dependence, appears to be a general metabolic defect in cancer. We have previously shown that cancer-cell growth can be selectively arrested by methionine deprivation such as with recombinant methioninase (rMETase). The present study used a previously-established patient-derived orthotopic xenograft (PDOX) nude mouse model of BRAF V600E-mutant melanoma to determine the efficacy of rMETase in combination with a first-line melanoma drug, temozolomide (TEM). In the present study 40 melanoma PDOX mouse models were randomized into four groups of 10 mice each: untreated control (n=10); TEM (25 mg/kg, oral 14 consecutive days, n=10); rMETase (100 units, intraperitoneal 14 consecutive days, n=10); combination TEM + rMETase (TEM: 25 mg/kg, oral rMETase: 100 units, intraperitoneal 14 consecutive days, n=10). All treatments inhibited tumor growth compared to untreated control (TEM: p =0.0081, rMETase: p =0.0037, TEM-rMETase: p =0.0024) on day 14 after initiation. However, the combination therapy of TEM and rMETase was significantly more efficacious than either mono-therapy (TEM: p =0.0051, rMETase: p =0.0051). The present study is the first demonstrating the efficacy of rMETase combination therapy in a PDOX model, suggesting potential clinical development, especially in recalcitrant cancers such as melanoma, where rMETase may enhance first-line therapy.
- Published
- 2017
- Full Text
- View/download PDF
11. Recombinant methioninase effectively targets a Ewing's sarcoma in a patient-derived orthotopic xenograft (PDOX) nude-mouse model
- Author
-
Qinghong Han, Takashi Murakami, Takashi Chishima, Arun S. Singh, Thinzar M. Lwin, Kentaro Igarashi, Kei Kawaguchi, Itaru Endo, Robert M. Hoffman, Fritz C. Eilber, Scott D. Nelson, Shukuan Li, Tasuku Kiyuna, Yukihiko Hiroshima, Ho Kyoung Hwang, Jonathan C. DeLong, Yuying Tan, Kentaro Miyake, Kuniya Tanaka, Yunfeng Li, Michael Bouvet, and Sarah M. Dry
- Subjects
0301 basic medicine ,Male ,Antimetabolites ,Biopsy ,Nude ,law.invention ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Nude mouse ,law ,patient-derived orthotopic xenograft ,Cancer ,methionine dependence ,Pediatric ,biology ,Sarcoma ,recombinant methioninase ,Ewing's sarcoma ,Antineoplastic ,Immunohistochemistry ,nude mice ,Recombinant Proteins ,3. Good health ,Tumor Burden ,Carbon-Sulfur Lyases ,Oncology ,030220 oncology & carcinogenesis ,Recombinant DNA ,Ewing’s sarcoma ,Biotechnology ,Research Paper ,medicine.medical_specialty ,Pediatric Research Initiative ,Antimetabolites, Antineoplastic ,Pediatric Cancer ,Oncology and Carcinogenesis ,Mice, Nude ,Sarcoma, Ewing ,03 medical and health sciences ,Rare Diseases ,Ewing ,medicine ,Animals ,Humans ,Methionine ,business.industry ,Animal ,medicine.disease ,biology.organism_classification ,Xenograft Model Antitumor Assays ,Surgery ,Brain Disorders ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Cell culture ,Disease Models ,Cancer research ,business ,recalcitrant cancer ,Transmethylation - Abstract
Methionine dependence is due to the overuse of methionine for aberrant transmethylation reactions in cancer. Methionine dependence may be the only general metabolic defect in cancer. In order to exploit methionine dependence for therapy, our laboratory previously cloned L-methionine α-deamino-γ-mercaptomethane lyase [EC 4.4.1.11]). The cloned methioninase, termed recombinant methioninase, or rMETase, has been tested in mouse models of human cancer cell lines. Ewing's sarcoma is recalcitrant disease even though development of multimodal therapy has improved patients'outcome. Here we report efficacy of rMETase against Ewing's sarcoma in a patient-derived orthotopic xenograft (PDOX) model. The Ewing's sarcoma was implanted in the right chest wall of nude mice to establish a PDOX model. Eight Ewing's sarcoma PDOX mice were randomized into untreated control group (n = 4) and rMETase treatment group (n = 4). rMETase (100 units) was injected intraperitoneally (i.p.) every 24 hours for 14 consecutive days. All mice were sacrificed on day-15, 24 hours after the last rMETase administration. rMETase effectively reduced tumor growth compared to untreated control. The methionine level both of plasma and supernatants derived from sonicated tumors was lower in the rMETase group. Body weight did not significantly differ at any time points between the 2 groups. The present study is the first demonstrating rMETase efficacy in a PDOX model, suggesting potential clinical development, especially in recalcitrant cancers such as Ewing's sarcoma.
- Published
- 2017
12. Efficacy of Recombinant Methioninase (rMETase) on Recalcitrant Cancer Patient-Derived Orthotopic Xenograft (PDOX) Mouse Models: A Review
- Author
-
Kentaro Igarashi, Robert M. Hoffman, Shukuan Li, Qinghong Han, Yuying Tan, Takashi Murakami, Michiaki Unno, and Kei Kawaguchi
- Subjects
0301 basic medicine ,Mice, Nude ,Review ,Methionine dependence ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Methionine ,law ,Neoplasms ,medicine ,Animals ,Humans ,Cytotoxicity ,lcsh:QH301-705.5 ,methionine dependence ,Cancer ,recombinant methioninase ,General Medicine ,medicine.disease ,Xenograft Model Antitumor Assays ,Recombinant Proteins ,nude mice ,Highly sensitive ,Carbon-Sulfur Lyases ,030104 developmental biology ,Treatment Outcome ,patient-derived tumor ,chemistry ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,Cancer cell ,Recombinant DNA ,Cancer research ,orthotopic implantation - Abstract
An excessive requirement for methionine (MET), termed MET dependence, appears to be a general metabolic defect in cancer and has been shown to be a very effective therapeutic target. MET restriction (MR) has inhibited the growth of all major cancer types by selectively arresting cancer cells in the late-S/G2 phase, when they also become highly sensitive to cytotoxic agents. Recombinant methioninase (rMETase) has been developed to effect MR. The present review describes the efficacy of rMETase on patient-derived orthotopic xenograft (PDOX) models of recalcitrant cancer, including the surprising result that rMETase administrated orally can be highly effective.
- Published
- 2019
13. HuR-mediated nucleocytoplasmic translocation of HOTAIR relieves its inhibition of osteogenic differentiation and promotes bone formation
- Author
-
Yuheng Li, Weijia Sun, Jianwei Li, Ruikai Du, Wenjuan Xing, Xinxin Yuan, Guohui Zhong, Dingsheng Zhao, Zizhong Liu, Xiaoyan Jin, Junjie Pan, Youyou Li, Qi Li, Guanghan Kan, Xuan Han, Shukuan Ling, Xiqing Sun, and Yingxian Li
- Subjects
Biology (General) ,QH301-705.5 ,Physiology ,QP1-981 - Abstract
Abstract Bone marrow mesenchymal stem cell (BMSC) osteogenic differentiation and osteoblast function play critical roles in bone formation, which is a highly regulated process. Long noncoding RNAs (lncRNAs) perform diverse functions in a variety of biological processes, including BMSC osteogenic differentiation. Although several studies have reported that HOX transcript antisense RNA (HOTAIR) is involved in BMSC osteogenic differentiation, its effect on bone formation in vivo remains unclear. Here, by constructing transgenic mice with BMSC (Prx1-HOTAIR)- and osteoblast (Bglap-HOTAIR)-specific overexpression of HOTAIR, we found that Prx1-HOTAIR and Bglap-HOTAIR transgenic mice show different bone phenotypes in vivo. Specifically, Prx1-HOTAIR mice showed delayed bone formation, while Bglap-HOTAIR mice showed increased bone formation. HOTAIR inhibits BMSC osteogenic differentiation but promotes osteoblast function in vitro. Furthermore, we identified that HOTAIR is mainly located in the nucleus of BMSCs and in the cytoplasm of osteoblasts. HOTAIR displays a nucleocytoplasmic translocation pattern during BMSC osteogenic differentiation. We first identified that the RNA-binding protein human antigen R (HuR) is responsible for HOTAIR nucleocytoplasmic translocation. HOTAIR is essential for osteoblast function, and cytoplasmic HOTAIR binds to miR-214 and acts as a ceRNA to increase Atf4 protein levels and osteoblast function. Bglap-HOTAIR mice, but not Prx1-HOTAIR mice, showed alleviation of bone loss induced by unloading. This study reveals the importance of temporal and spatial regulation of HOTAIR in BMSC osteogenic differentiation and bone formation, which provides new insights into precise regulation as a target for bone loss.
- Published
- 2023
- Full Text
- View/download PDF
14. High Efficacy of Recombinant Methioninase on Patient-Derived Orthotopic Xenograft (PDOX) Mouse Models of Cancer
- Author
-
Robert M, Hoffman, Takashi, Murakami, Kei, Kawaguchi, Kentaro, Igarashi, Yuying, Tan, Shukuan, Li, and Qinghong, Han
- Subjects
Proto-Oncogene Proteins B-raf ,Carbon-Sulfur Lyases ,Time Factors ,Neoplasms ,Body Weight ,Mutation ,Temozolomide ,Animals ,Humans ,Mice, Nude ,Xenograft Model Antitumor Assays ,Recombinant Proteins ,Cell Proliferation - Abstract
Methionine (MET) is a general target in cancer due to the excess requirement of MET by cancer cells. MET has been effectively restricted by recombinant methioninase (rMETase) in mouse models of cell-line tumors. This chapter reviews the efficacy of rMETase on patient-derived orthotopic xenograft (PDOX) mouse models of human cancer. Ewing's sarcoma is a recalcitrant disease even though development of multimodal therapy has improved patients' outcome. A Ewing's sarcoma was implanted in the right chest wall of nude mice to establish a PDOX model. rMETase effectively reduced tumor growth compared to the untreated control. The MET level both of plasma and supernatants derived from sonicated tumors was lower in the rMETase treatment group. Body weight did not significantly differ at any time points between the two groups. A PDOX nude mouse model of a BRAF V600E-mutant melanoma was established in the chest wall of nude mice and also tested with rMETase in combination with a first-line melanoma drug, temozolomide (TEM). Combination therapy of TEM and rMETase was significantly more efficacious than either monotherapy. The results reviewed in this chapter demonstrate the clinical potential of rMETase.
- Published
- 2019
15. Safety and Toxicity of Recombinant Methioninase and Polyethylene Glycol (PEG) Recombinant Methioninase in Primates
- Author
-
Robert M, Hoffman, Zhijian, Yang, Yuying, Tan, Qinghong, Han, Shukuan, Li, and Shigeo, Yagi
- Subjects
Primates ,Carbon-Sulfur Lyases ,Mice ,Methionine ,Animals ,Xenograft Model Antitumor Assays ,Antibodies ,Recombinant Proteins ,Polyethylene Glycols - Abstract
Methionine (MET) is a general metabolic therapeutic target in cancer, whereby cancer cells have an elevated requirement for MET, termed MET dependence. We have developed recombinant L-methionine α-deamino-γ-mercaptomethane lyase (recombinant methioninase [rMETase, EC 4.4.1.11]) as targeted therapy of all cancer types. Pharmacokinetics, MET depletion, antigenicity, and toxicity of rMETase were examined in macaque monkeys. Pharmacokinetic analysis showed that rMETase was eliminated with a T
- Published
- 2019
16. Afterword: Oral Methioninase-Answer to Cancer and Fountain of Youth?
- Author
-
Robert M, Hoffman, Qinghong, Han, Kei, Kawaguchi, Shukuan, Li, and Yuying, Tan
- Subjects
Aging ,Carbon-Sulfur Lyases ,Neoplasms ,Administration, Oral ,Animals ,Humans ,Mice, Nude ,Female ,Xenograft Model Antitumor Assays ,Recombinant Proteins ,Aged - Abstract
The elevated methionine (MET) requirement of cancer cells is termed MET dependence and is possibly the only known general metabolic defect in cancer. Targeting MET by recombinant methioninase (rMETase) can arrest the growth of cancer cells in vitro and in vivo due to their elevated requirement for MET. rMETase can also potentiate chemotherapy drugs active in S phase due to the selective arrest of cancer cells in S/G
- Published
- 2019
17. Development of Recombinant Methioninase for Cancer Treatment
- Author
-
Robert M, Hoffman, Yuying, Tan, Shukuan, Li, Qinghong, Han, Shigeo, Yagi, Tomoaki, Takakura, Akio, Takimoto, Kenji, Inagaki, and Daizou, Kudou
- Subjects
Carbon-Sulfur Lyases ,Mice, Inbred BALB C ,Apoenzymes ,Pseudomonas putida ,Neoplasms ,Pyridoxal Phosphate ,Fermentation ,Escherichia coli ,Animals ,Crystallization ,Recombinant Proteins ,Polyethylene Glycols - Abstract
The elevated requirement for methionine (MET) of cancer cells is termed MET dependence. To selectively target the MET dependence of tumors for treatment on a large-scale preclinical and clinical basis, the L-methionine α-deamino-γ-mercaptomethane-lyase (EC 4.4.1.11) (methioninase, [METase]) gene from Pseudomonas putida has been cloned in Escherichia coli using the polymerase chain reaction (PCR). Purification using two DEAE Sepharose FF ion-exchange column and one ActiClean Etox endotoxin-affinity chromatography column has been established. Plasmid pMGLTrc03, which has a trc promoter and a spacing of 12 nucleotides between the Shine-Dalgarno sequence and the ATG translation initiation codon, was selected as the most suitable plasmid. The recombinant bacteria produced rMETase at 43% of the total proteins in soluble fraction by simple batch fermentation using a 500 L fermentor. Crystals were directly obtained from crude enzyme with 87% yield by a crystallization in the presence of 9.0% polyethylene glycol 6000, 3.6% ammonium sulfate, and 0.18 M sodium chloride using a 100 L crystallizer. After recrystallization, the enzyme was purified by anion-exchange column chromatography to remove endotoxins and by gel filtration for polishing. Purified rMETase is stable to lyophilization. In order to prevent immunological reactions which might be produced by multiple dosing of rMETase and to prolong the serum half-life of rMETase, the N-hydroxysuccinimidyl ester of methoxypolyethylene glycol propionic acid (M-SPA-PEG 5000) has been coupled to rMETase. The PEGylated molecules (PEG-rMETase) were purified from unreacted PEG with Amicon 30 K centriprep concentrators or by Sephacryl S-300 HR gel-filtration chromatography. Unreacted rMETase was removed by DEAE Sepharose FF anion-exchange chromatography. The resulting PEG-rMETase subunit, produced from a PEG/rMETase ratio of 30/1 in the synthetic reaction, had a molecular mass of approximately 53 kda determined by matrix-assisted laser desorption/ionization mass spectrometry, indicating the conjugation of two PEG molecules per subunit of rMETase and eight per tetramer. PEG-rMETase molecules obtained from reacting ratios of PEG/rMETase of 30/1 had an enzyme activity of 70% of unmodified rMETase. PEGylation of rMETase increased the serum half-life of the enzyme in rats to approximately 160 min compared to 80 min for unmodified rMETase. PEG-rMETase could deplete serum MET levels to less than 0.1 μM for approximately 8 h compared to 2 h for rMETase in rats. A significant prolongation of in vivo activity and effective MET depletion by the PEG-rMETase were achieved by the simultaneous administration of pyridoxal 5'-phosphate. rMETase was also conjugated with methoxypolyethylene glycol succinimidyl glutarate 5000 (MEGC-PEG). Miniosmotic pumps containing various concentrations of PLP were implanted in BALB-C mice. PLP-infused mice were then injected with a single dose of 4000 or 8000 units/kg PEG-rMETase. Mice infused with 5, 50, 100, 200, and 500 mg/mL PLP-containing miniosmotic pumps increased plasma PLP to 7, 24, 34, 60, and 95 μM, respectively, from the PLP baseline of 0.3 μM. PLP increased the half-life of MEGC-PEG-rMETase holoenzyme in a dose-dependent manner. The extended time of MET depletion by MEGC-PEG-rMETase was due to the maintenance of active MEGC-PEG-rMETase holoenzyme by infused PLP.
- Published
- 2019
18. Pilot Phase I Clinical Trial of Methioninase on High-Stage Cancer Patients: Rapid Depletion of Circulating Methionine
- Author
-
Robert M, Hoffman, Yuying, Tan, Shukuan, Li, Qinghong, Han, and Jorge, Zavala
- Subjects
Carbon-Sulfur Lyases ,Methionine ,Neoplasms ,Fermentation ,Humans ,Pilot Projects ,Infusions, Intravenous ,Neoplasm Staging - Abstract
Methionine (MET) has been shown to be a tumor-selective therapeutic target for cancer, since cancer cells require higher amounts of MET to divide and survive than normal cells. This phenomena is known as MET dependence and is probably due to MET overuse by cancer cells. A pilot clinical trial was initially carried out with non-recombinant METase (METase) produced from Pseudomonas putida and subsequently highly purified. No acute clinical toxicity was observed for any criteria measured in the three patients. The depletion of serum MET started within 30 min of the infusion and was maintained for 4 h after the infusion was completed in patient 1 and patient 2. The lowest serum MET levels were 35% and 19% of the pretreatment level, respectively, in patient 1 and patient 2. Patient 3 received a 10 h i.v. infusion of METase without any sign of side effects. MET was depleted over 200-fold from 23.1 to 0.1 μM by the 10-h infusion of patient 3. No clinical toxicity was observed in any criteria measured in patient 3. Subsequently, another pilot Phase I clinical trial was carried out of serum MET depletion in cancer patients by recombinant METase (rMETase) cloned from Pseudomonas putida and produced in E. coli. Patients with advanced breast cancer, lung cancer, renal cancer, and lymphoma were given a single rMETase treatment at doses ranging from 5000 to 20,000 units by i.v. infusion over 6-24 h. No clinical toxicity was observed in any patient after rMETase treatment. rMETase levels were measured at 0.1 to 0.4 units per ml of serum in the patients which correspond to therapeutic levels in vitro. The lowest serum MET levels in rMETase-treated patients were 0.1% of the pretreatment levels corresponding to approximately 0.1 μM, which also correlates to therapeutic levels in vitro as well as in vivo. The results of the METase and rMETase pilot Phase I clinical trials therefore indicate that i.v. infusion of rMETase is safe and effectively depletes its biochemical target of serum MET, suggesting potential efficacy in future clinical trials.
- Published
- 2019
19. Safety and Toxicity of Recombinant Methioninase and Polyethylene Glycol (PEG) Recombinant Methioninase in Primates
- Author
-
Shukuan Li, Shigeo Yagi, Zhijian Yang, Robert M. Hoffman, Yuying Tan, and Qinghong Han
- Subjects
0301 basic medicine ,Antigenicity ,Methionine ,biology ,Chemistry ,Serum albumin ,Pharmacology ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Pharmacokinetics ,law ,030220 oncology & carcinogenesis ,PEG ratio ,Toxicity ,biology.protein ,Recombinant DNA ,Antibody - Abstract
Methionine (MET) is a general metabolic therapeutic target in cancer, whereby cancer cells have an elevated requirement for MET, termed MET dependence. We have developed recombinant L-methionine α-deamino-γ-mercaptomethane lyase (recombinant methioninase [rMETase, EC 4.4.1.11]) as targeted therapy of all cancer types. Pharmacokinetics, MET depletion, antigenicity, and toxicity of rMETase were examined in macaque monkeys. Pharmacokinetic analysis showed that rMETase was eliminated with a T1/2 of 2.49 h. A 2-week i.v. administration of 4000 units/kg every 8 h/day for 2 weeks resulted in a steady-state depletion of plasma MET to less than 2 μM. The only manifest toxicity was decreased food intake and slight weight loss. Serum albumin and red-cell values declined transiently during treatment. Rechallenge on day 28 resulted in anaphylactic shock and death in one animal. Pretreatment with hydrocortisone prevented the anaphylactic reaction. Anti-rMETase antibodies (at 10-3) were found after the first challenge, increased to 10-6 after the fourth challenge, and decreased to 10-2 by 2 months post-therapy. Therefore, the therapeutic potential of rMETase is limited by its short plasma half-life and immunologic effects, including high antibody production in mice and anaphylactic reactions in monkeys. To overcome these limits, rMETase has been coupled to methoxypolyethylene glycol succinimidyl glutarate polyethylene glycol (MEGC-PEG-5000). The pharmacokinetics, antigenicity, and toxicity of MEGC-PEG-rMETase in macaque monkeys were evaluated using an escalating-dose strategy. In pharmacokinetic studies, a single 4000 units/kg dose showed that MEGC-PEG-rMETase holoenzyme activity was eliminated with a biological half-life of 1.3 h, and the MEGC-PEG-rMETase apoenzyme was eliminated with a biological half-life of 90 h, a 36-fold increase compared with non-PEGylated rMETase. The disparity in the T½ of the apoenzyme and the holoenzyme reflects the loss of co-factor pyridoxal-L-phosphate of the circulating MEGC-PEG-rMETase. A 7-day i.v. administration of 4000 units/kg every 12 h resulted in a steady-state depletion of plasma MET to
- Published
- 2019
- Full Text
- View/download PDF
20. Pilot Phase I Clinical Trial of Methioninase on High-Stage Cancer Patients: Rapid Depletion of Circulating Methionine
- Author
-
Robert M. Hoffman, Yuying Tan, Shukuan Li, Qinghong Han, and Jorge Zavala
- Subjects
0301 basic medicine ,Methionine ,business.industry ,Cancer ,Pharmacology ,medicine.disease ,Lymphoma ,Clinical trial ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,In vivo ,030220 oncology & carcinogenesis ,Cancer cell ,Toxicity ,medicine ,business ,Lung cancer - Abstract
Methionine (MET) has been shown to be a tumor-selective therapeutic target for cancer, since cancer cells require higher amounts of MET to divide and survive than normal cells. This phenomena is known as MET dependence and is probably due to MET overuse by cancer cells. A pilot clinical trial was initially carried out with non-recombinant METase (METase) produced from Pseudomonas putida and subsequently highly purified. No acute clinical toxicity was observed for any criteria measured in the three patients. The depletion of serum MET started within 30 min of the infusion and was maintained for 4 h after the infusion was completed in patient 1 and patient 2. The lowest serum MET levels were 35% and 19% of the pretreatment level, respectively, in patient 1 and patient 2. Patient 3 received a 10 h i.v. infusion of METase without any sign of side effects. MET was depleted over 200-fold from 23.1 to 0.1 μM by the 10-h infusion of patient 3. No clinical toxicity was observed in any criteria measured in patient 3. Subsequently, another pilot Phase I clinical trial was carried out of serum MET depletion in cancer patients by recombinant METase (rMETase) cloned from Pseudomonas putida and produced in E. coli. Patients with advanced breast cancer, lung cancer, renal cancer, and lymphoma were given a single rMETase treatment at doses ranging from 5000 to 20,000 units by i.v. infusion over 6-24 h. No clinical toxicity was observed in any patient after rMETase treatment. rMETase levels were measured at 0.1 to 0.4 units per ml of serum in the patients which correspond to therapeutic levels in vitro. The lowest serum MET levels in rMETase-treated patients were 0.1% of the pretreatment levels corresponding to approximately 0.1 μM, which also correlates to therapeutic levels in vitro as well as in vivo. The results of the METase and rMETase pilot Phase I clinical trials therefore indicate that i.v. infusion of rMETase is safe and effectively depletes its biochemical target of serum MET, suggesting potential efficacy in future clinical trials.
- Published
- 2019
- Full Text
- View/download PDF
21. Development of Recombinant Methioninase for Cancer Treatment
- Author
-
Shigeo Yagi, Kenji Inagaki, Qinghong Han, Akio Takimoto, Tomoaki Takakura, Shukuan Li, Robert M. Hoffman, Daizou Kudou, and Yuying Tan
- Subjects
0301 basic medicine ,Chromatography ,Molecular mass ,Size-exclusion chromatography ,Polyethylene glycol ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Column chromatography ,DEAE-Sepharose ,chemistry ,030220 oncology & carcinogenesis ,PEG ratio ,PEGylation ,Chromatography column - Abstract
The elevated requirement for methionine (MET) of cancer cells is termed MET dependence. To selectively target the MET dependence of tumors for treatment on a large-scale preclinical and clinical basis, the L-methionine α-deamino-γ-mercaptomethane-lyase (EC 4.4.1.11) (methioninase, [METase]) gene from Pseudomonas putida has been cloned in Escherichia coli using the polymerase chain reaction (PCR). Purification using two DEAE Sepharose FF ion-exchange column and one ActiClean Etox endotoxin-affinity chromatography column has been established. Plasmid pMGLTrc03, which has a trc promoter and a spacing of 12 nucleotides between the Shine-Dalgarno sequence and the ATG translation initiation codon, was selected as the most suitable plasmid. The recombinant bacteria produced rMETase at 43% of the total proteins in soluble fraction by simple batch fermentation using a 500 L fermentor. Crystals were directly obtained from crude enzyme with 87% yield by a crystallization in the presence of 9.0% polyethylene glycol 6000, 3.6% ammonium sulfate, and 0.18 M sodium chloride using a 100 L crystallizer. After recrystallization, the enzyme was purified by anion-exchange column chromatography to remove endotoxins and by gel filtration for polishing. Purified rMETase is stable to lyophilization. In order to prevent immunological reactions which might be produced by multiple dosing of rMETase and to prolong the serum half-life of rMETase, the N-hydroxysuccinimidyl ester of methoxypolyethylene glycol propionic acid (M-SPA-PEG 5000) has been coupled to rMETase. The PEGylated molecules (PEG-rMETase) were purified from unreacted PEG with Amicon 30 K centriprep concentrators or by Sephacryl S-300 HR gel-filtration chromatography. Unreacted rMETase was removed by DEAE Sepharose FF anion-exchange chromatography. The resulting PEG-rMETase subunit, produced from a PEG/rMETase ratio of 30/1 in the synthetic reaction, had a molecular mass of approximately 53 kda determined by matrix-assisted laser desorption/ionization mass spectrometry, indicating the conjugation of two PEG molecules per subunit of rMETase and eight per tetramer. PEG-rMETase molecules obtained from reacting ratios of PEG/rMETase of 30/1 had an enzyme activity of 70% of unmodified rMETase. PEGylation of rMETase increased the serum half-life of the enzyme in rats to approximately 160 min compared to 80 min for unmodified rMETase. PEG-rMETase could deplete serum MET levels to less than 0.1 μM for approximately 8 h compared to 2 h for rMETase in rats. A significant prolongation of in vivo activity and effective MET depletion by the PEG-rMETase were achieved by the simultaneous administration of pyridoxal 5'-phosphate. rMETase was also conjugated with methoxypolyethylene glycol succinimidyl glutarate 5000 (MEGC-PEG). Miniosmotic pumps containing various concentrations of PLP were implanted in BALB-C mice. PLP-infused mice were then injected with a single dose of 4000 or 8000 units/kg PEG-rMETase. Mice infused with 5, 50, 100, 200, and 500 mg/mL PLP-containing miniosmotic pumps increased plasma PLP to 7, 24, 34, 60, and 95 μM, respectively, from the PLP baseline of 0.3 μM. PLP increased the half-life of MEGC-PEG-rMETase holoenzyme in a dose-dependent manner. The extended time of MET depletion by MEGC-PEG-rMETase was due to the maintenance of active MEGC-PEG-rMETase holoenzyme by infused PLP.
- Published
- 2019
- Full Text
- View/download PDF
22. Afterword: Oral Methioninase—Answer to Cancer and Fountain of Youth?
- Author
-
Shukuan Li, Robert M. Hoffman, Yuying Tan, Qinghong Han, and Kei Kawaguchi
- Subjects
0301 basic medicine ,Cancer prevention ,business.industry ,medicine.medical_treatment ,Melanoma ,Intraperitoneal injection ,Cancer ,medicine.disease ,In vitro ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,Cancer cell ,Toxicity ,medicine ,Cancer research ,business - Abstract
The elevated methionine (MET) requirement of cancer cells is termed MET dependence and is possibly the only known general metabolic defect in cancer. Targeting MET by recombinant methioninase (rMETase) can arrest the growth of cancer cells in vitro and in vivo due to their elevated requirement for MET. rMETase can also potentiate chemotherapy drugs active in S phase due to the selective arrest of cancer cells in S/G2 phase during MET restriction (MR). We previously reported that rMETase, administrated by intraperitoneal injection (ip-rMETase), could inhibit tumor growth in mouse models of cancer including patient-derived orthotopic xenograft (PDOX) mouse models. We subsequently compared ip-rMETase and oral rMETase (o-rMETase) on a melanoma PDOX mouse model. o-rMETase was significantly more effective than ip-rMETase to inhibit tumor growth without overt toxicity. The combination of o-rMETase+ip-rMETase was significantly more effective than either monotherapy and completely arrested tumor growth. Thus, o-rMETase is effective as an anticancer agent with the potential of clinical development for chronic cancer therapy as well as for cancer prevention. o-rMETase may also have potential as an antiaging agent for healthy people, since MR has been shown to extend the life span of a variety of different organisms.
- Published
- 2019
- Full Text
- View/download PDF
23. High Efficacy of Recombinant Methioninase on Patient-Derived Orthotopic Xenograft (PDOX) Mouse Models of Cancer
- Author
-
Kentaro Igarashi, Yuying Tan, Kei Kawaguchi, Robert M. Hoffman, Shukuan Li, Qinghong Han, and Takashi Murakami
- Subjects
0301 basic medicine ,Chemotherapy ,Temozolomide ,Combination therapy ,biology ,business.industry ,Melanoma ,medicine.medical_treatment ,Cancer ,medicine.disease ,biology.organism_classification ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Nude mouse ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,medicine ,Sarcoma ,business ,medicine.drug - Abstract
Methionine (MET) is a general target in cancer due to the excess requirement of MET by cancer cells. MET has been effectively restricted by recombinant methioninase (rMETase) in mouse models of cell-line tumors. This chapter reviews the efficacy of rMETase on patient-derived orthotopic xenograft (PDOX) mouse models of human cancer. Ewing's sarcoma is a recalcitrant disease even though development of multimodal therapy has improved patients' outcome. A Ewing's sarcoma was implanted in the right chest wall of nude mice to establish a PDOX model. rMETase effectively reduced tumor growth compared to the untreated control. The MET level both of plasma and supernatants derived from sonicated tumors was lower in the rMETase treatment group. Body weight did not significantly differ at any time points between the two groups. A PDOX nude mouse model of a BRAF V600E-mutant melanoma was established in the chest wall of nude mice and also tested with rMETase in combination with a first-line melanoma drug, temozolomide (TEM). Combination therapy of TEM and rMETase was significantly more efficacious than either monotherapy. The results reviewed in this chapter demonstrate the clinical potential of rMETase.
- Published
- 2019
- Full Text
- View/download PDF
24. Combining Tumor-Selective Bacterial Therapy with Salmonella typhimurium A1-R and Cancer Metabolism Targeting with Oral Recombinant Methioninase Regressed an Ewing's Sarcoma in a Patient-Derived Orthotopic Xenograft Model
- Author
-
Kentaro, Miyake, Tasuku, Kiyuna, Shukuan, Li, Qinghong, Han, Yuying, Tan, Ming, Zhao, Hiromichi, Oshiro, Kei, Kawaguchi, Takashi, Higuchi, Zhiying, Zhang, Sahar, Razmjooei, Maryam, Barangi, Sintawat, Wangsiricharoen, Takashi, Murakami, Arun S, Singh, Yunfeng, Li, Scott D, Nelson, Fritz C, Eilber, Michael, Bouvet, Yukihiko, Hiroshima, Takashi, Chishima, Ryusei, Matsuyama, Shree Ram, Singh, Itaru, Endo, and Robert M, Hoffman
- Subjects
Salmonella typhimurium ,Antibiotics, Antineoplastic ,Body Weight ,Transplantation, Heterologous ,Administration, Oral ,Mice, Nude ,Bone Neoplasms ,Sarcoma, Ewing ,Recombinant Proteins ,Carbon-Sulfur Lyases ,Disease Models, Animal ,Mice ,Doxorubicin ,Animals ,Humans ,Female - Abstract
Ewing's sarcoma (ES) is a recalcitrant disease in need of transformative therapeutics.The aim of this study was to investigate the efficacy of tumor-selective Salmonella typhimurium A1-R combined with tumor metabolism targeting with oral administration of recombinant methioninase (o-rMETase), on an ES patient-derived orthotopic xenograft (PDOX) model.The ES PDOX models were previously established in the right chest wall. The ES PDOX models were randomized into 5 groups when the tumor volume reached 80 mm3: G1: untreated control; G2: doxorubicin; G3: S. typhimurium A1-R; G4: o-rMETase; G5: S. typhimurium A1-R combined with o-rMETase. All mice were sacrificed on day 15. Body weight and tumor volume were assessed twice a week.S. typhimurium A1-R and o-rMETase respectively suppressed tumor growth as monotherapies (p = 0.050 and p = 0.032). S. typhimurium A1-R combined with o-rMETase regressed tumor growth significantly compared to untreated group on day 15 (p0.032). S. typhimurium A1-R combined with o-rMETase group was significantly more effective than S. typhimurium A1-R or o-rMETase monotherapy (p = 0.032, p = 0.032).The present results suggest that the combination of S. typhimurium A1-R and o-rMETase has promise to be a transformative therapy for ES.
- Published
- 2018
25. Combination therapy of tumor-targeting Salmonella typhimurium A1-R and oral recombinant methioninase regresses a BRAF-V600E-negative melanoma
- Author
-
Norihiko Sugisawa, Yunfeng Li, Sahar Razmjooei, Shukuan Li, Scott D. Nelson, Robert M. Hoffman, Sarah M. Dry, Hiromichi Ohshiro, Mark A. Eckardt, Kentaro Miyake, Sintawat Wangsiricharoen, Fritz C. Eilber, Zhiying Zhang, Qinghong Han, Arun S. Singh, Shree Ram Singh, Michiaki Unno, Takashi Higuchi, Kei Kawaguchi, Yuying Tan, Masuyo Miyake, Ming Zhao, Kentaro Igarashi, Tasuku Kiyuna, Bartosz Chmielowski, and Tara A. Russell
- Subjects
0301 basic medicine ,Male ,Proto-Oncogene Proteins B-raf ,Salmonella typhimurium ,Salmonella ,Tumor targeting ,Antimetabolites, Antineoplastic ,Combination therapy ,Biophysics ,Administration, Oral ,Mice, Nude ,Antineoplastic Agents ,medicine.disease_cause ,Biochemistry ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Drug Delivery Systems ,law ,medicine ,Temozolomide ,Animals ,Humans ,Point Mutation ,Molecular Biology ,Melanoma ,business.industry ,Pseudomonas putida ,Cancer ,Cell Biology ,medicine.disease ,Recombinant Proteins ,BRAF V600E ,Carbon-Sulfur Lyases ,Disease Models, Animal ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Recombinant DNA ,business ,medicine.drug - Abstract
Melanoma is a recalcitrant cancer. To improve and individualize treatment for this disease, we previously developed a patient-derived orthotopic xenograft (PDOX) model for melanoma. We previously reported the individual efficacy of tumor-targeting Salmonella typhimurium A1-R (S. typhimurium A1-R) and recombinant methioninase (rMETase) for melanoma in the PDOX models of this disease. In the present study, we evaluated the efficacy of the combination of S. typhimurium A1-R with orally-administered rMETase (o-rMETase) for BRAF-V600E-negative melanoma in a PDOX model. Three weeks after implantation, 60 PDOX mouse models were randomized into six groups of 10 mice each: untreated control, temozolomide (TEM); o-rMETase; S. typhimurium A1-R; TEM + rMETase, S. typhimurium A1-R + rMETase. All treatments inhibited tumor growth compared to untreated control (TEM: p
- Published
- 2018
26. Tumor-targeting Salmonella typhimurium A1-R combined with recombinant methioninase and cisplatinum eradicates an osteosarcoma cisplatinum-resistant lung metastasis in a patient-derived orthotopic xenograft (PDOX) mouse model: decoy, trap and kill chemotherapy moves toward the clinic
- Author
-
Sarah M. Dry, Mark A. Eckardt, Irmina A. Elliott, Masuyo Miyake, Kentaro Igarashi, Norio Yamamoto, Arun S. Singh, Yunfeng Li, Ming Zhao, Shukuan Li, Hiroyuki Tsuchiya, Qinghong Han, Scott D. Nelson, Fritz C. Eilber, Shinji Miwa, Hiroaki Kimura, Tara A. Russell, Kentaro Miyake, Kei Kawaguchi, Yuying Tan, Katsuhiro Hayashi, Robert M. Hoffman, and Tasuku Kiyuna
- Subjects
0301 basic medicine ,Salmonella typhimurium ,Salmonella ,Lung Neoplasms ,medicine.medical_treatment ,Nude ,Lung metastasis ,Drug Resistance ,medicine.disease_cause ,law.invention ,Metastasis ,Mice ,0302 clinical medicine ,cisplatinum ,law ,Tumor Cells, Cultured ,Salmonella typhimurium A1R ,Cancer ,Heterologous ,Osteosarcoma ,Cultured ,Recombinant Proteins ,Tumor Cells ,G2 Phase Cell Cycle Checkpoints ,Carbon-Sulfur Lyases ,Local ,030220 oncology & carcinogenesis ,Combination ,S Phase Cell Cycle Checkpoints ,Recombinant DNA ,Drug Therapy, Combination ,Decoy ,trap ,Transplantation, Heterologous ,Mice, Nude ,Antineoplastic Agents ,Biology ,Salmonella typhimurium A1-R ,lung ,resistance ,PDOX ,03 medical and health sciences ,Rare Diseases ,Drug Therapy ,osteosarcoma ,medicine ,metastasis ,Animals ,Humans ,decoy ,Molecular Biology ,methioninase ,Transplantation ,Chemotherapy ,kill ,Animal ,Cell Biology ,medicine.disease ,Disease Models, Animal ,Neoplasm Recurrence ,030104 developmental biology ,Drug Resistance, Neoplasm ,Disease Models ,Cancer cell ,Cancer research ,Neoplasm ,Biochemistry and Cell Biology ,Cisplatin ,Neoplasm Recurrence, Local ,Developmental Biology ,Reports - Abstract
In the present study, a patient-derived orthotopic xenograft (PDOX) model of recurrent cisplatinum (CDDP)-resistant metastatic osteosarcoma was treated with Salmonella typhimurium A1-R (S. typhimurium A1-R), which decoys chemoresistant quiescent cancer cells to cycle, and recombinant methioninase (rMETase), which selectively traps cancer cells in late S/G2, and chemotherapy. The PDOX models were randomized into the following groups 14days after implantation: G1, control without treatment; G2, CDDP (6mg/kg, intraperitoneal (i.p.) injection, weekly, for 2weeks); G3, rMETase (100unit/mouse, i.p., daily, for 2weeks). G4, S. typhimurium A1-R (5× 107 CFU/100μl, i.v., weekly, for 2weeks); G5, S. typhimurium A1-R (5× 107 CFU/100μl, i.v., weekly, for 2weeks) combined with rMETase (100unit/mouse, i.p., daily, for 2weeks); G6, S. typhimurium A1-R (5× 107 CFU/100μl, i.v., weekly, for 2weeks) combined with rMETase (100unit/mouse, i.p., daily, for 2weeks) and CDDP (6mg/kg, i.p. injection, weekly, for 2weeks). On day 14 after initiation, all treatments except CDDP alone, significantly inhibited tumor growth compared to untreated control: (CDDP: p = 0.586; rMETase: p = 0.002; S. typhimurium A1-R: p = 0.002; S. typhimurium A1-R combined with rMETase: p = 0.0004; rMETase combined with both S. typhimurium A1-R and CDDP: p = 0.0001). The decoy, trap and kill combination of S. typhimurium A1-R, rMETase and CDDP was the most effective of all therapies and was able to eradicate the metastatic osteosarcoma PDOX.
- Published
- 2018
27. Targeting altered cancer methionine metabolism with recombinant methioninase (rMETase) overcomes partial gemcitabine-resistance and regresses a patient-derived orthotopic xenograft (PDOX) nude mouse model of pancreatic cancer
- Author
-
Hiromichi Oshiro, Kentaro Miyake, Michael Bouvet, Michiaki Unno, Shukuan Li, Kei Kawaguchi, Qinghong Han, Masuyo Miyake, Thinzar M. Lwin, Takashi Higuchi, Robert M. Hoffman, Kentaro Igarashi, Yuying Tan, and Tasuku Kiyuna
- Subjects
0301 basic medicine ,Male ,endocrine system diseases ,Antimetabolites ,Nude ,pancreatic cancer ,precision therapy ,Drug Resistance ,Gene Expression ,Deoxycytidine ,law.invention ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Nude mouse ,Methionine ,law ,Antineoplastic Combined Chemotherapy Protocols ,Molecular Targeted Therapy ,patient-derived orthotopic xenograft ,Cancer ,methionine dependence ,biology ,gemcitabine ,Antineoplastic ,nude mice ,Recombinant Proteins ,Tumor Burden ,Carbon-Sulfur Lyases ,030220 oncology & carcinogenesis ,Recombinant DNA ,Injections, Intraperitoneal ,medicine.drug ,Antimetabolites, Antineoplastic ,Combination therapy ,Methionine metabolism ,Mice, Nude ,Drug Administration Schedule ,Injections ,03 medical and health sciences ,Rare Diseases ,Pancreatic cancer ,Report ,medicine ,Animals ,Humans ,Intraperitoneal ,Molecular Biology ,orthotopic ,Recombinant methioninase ,Cell Biology ,medicine.disease ,biology.organism_classification ,Xenograft Model Antitumor Assays ,Gemcitabine ,Pancreatic Neoplasms ,030104 developmental biology ,Orphan Drug ,chemistry ,Drug Resistance, Neoplasm ,Cancer research ,Neoplasm ,Biochemistry and Cell Biology ,Digestive Diseases ,Developmental Biology - Abstract
Pancreatic cancer is a recalcitrant disease. Gemcitabine (GEM) is the most widely-used first-line therapy for pancreatic cancer, but most patients eventually fail. Transformative therapy is necessary to significantly improve the outcome of pancreatic cancer patients. Tumors have an elevated requirement for methionine and are susceptible to methionine restriction. The present study used a patient-derived orthotopic xenograft (PDOX) nude mouse model of pancreatic cancer to determine the efficacy of recombinant methioninase (rMETase) to effect methionine restriction and thereby overcome GEM-resistance. A pancreatic cancer obtained from a patient was grown orthotopically in the pancreatic tail of nude mice to establish the PDOX model. Five weeks after implantation, 40 pancreatic cancer PDOX mouse models were randomized into four groups of 10 mice each: untreated control (n = 10); GEM (100 mg/kg, i.p., once a week for 5 weeks, n = 10); rMETase (100 units, i.p., 14 consecutive days, n = 10); GEM+rMETase (GEM: 100 mg/kg, i.p., once a week for 5 weeks, rMETase: 100 units, i.p., 14 consecutive days, n = 10). Although GEM partially inhibited PDOX tumor growth, combination therapy (GEM+rMETase) was significantly more effective than mono therapy (GEM: p = 0.0025, rMETase: p = 0.0010). The present study is the first demonstrating the efficacy of rMETase combination therapy in a pancreatic cancer PDOX model to overcome first-line therapy resistance in this recalcitrant disease.
- Published
- 2018
28. Growth of doxorubicin-resistant undifferentiated spindle-cell sarcoma PDOX is arrested by metabolic targeting with recombinant methioninase
- Author
-
Tasuku Kiyuna, Hiroyuki Tsuchiya, Kentaro Miyake, Arun S. Singh, Sarah M. Dry, Scott D. Nelson, Mark A. Eckardt, Qinghong Han, Kei Kawaguchi, Takashi Murakami, Irmina A. Elliott, Katsuhiro Hayashi, Kentaro Igarashi, Shukuan Li, Yuying Tan, Tara A. Russell, Robert M. Hoffman, Yunfeng Li, Norio Yamamoto, Fritz C. Eilber, Shinji Miwa, and Hiroaki Kimura
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Nude ,Medical Physiology ,Docetaxel ,Biochemistry ,Deoxycytidine ,Mice ,0302 clinical medicine ,Medicine ,patient-derived orthotopic xenograft ,Melanoma ,Cancer ,Sulfonamides ,Sarcoma ,recombinant methioninase ,Carbon-Sulfur Lyases ,resistant ,030220 oncology & carcinogenesis ,Taxoids ,Female ,medicine.drug ,Biotechnology ,Biochemistry & Molecular Biology ,Indazoles ,Mice, Nude ,Sarcoma, Ewing ,doxorubicin ,Pazopanib ,03 medical and health sciences ,PDOX ,Rare Diseases ,Ewing ,Animals ,Doxorubicin ,Molecular Biology ,Chemotherapy ,business.industry ,Animal ,undifferentiated spindle-cell sarcoma ,Cell Biology ,medicine.disease ,Gemcitabine ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,Orphan Drug ,Pyrimidines ,Immunology ,Disease Models ,Cancer research ,Biochemistry and Cell Biology ,business - Abstract
Undifferentiated spindle-cell sarcoma (USCS) is a recalcitrant -cancer in need of individualized therapy. A high-grade USCS from a striated muscle of a patient was grown orthotopically in the right biceps femoris muscle of nude mice to establish a patient-derived orthotopic xenograft (PDOX) model. In a previous study, we evaluated the efficacy of standard first-line chemotherapy of doxorubicin (DOX), gemcitabine (GEM) combined with docetaxel (DOC), compared to pazopanib (PAZ), a multi-targeting tyrosine-kinase inhibitor, in an USCS PDOX model. In the present study, mice-bearing the USCS PDOX tumors were randomized into the following groups when tumor volume reached 100 mm3 : G1, untreated control without treatment; G2, DOX (3 mg/kg, intraperitoneal (i.p.) injection, weekly, for 2 weeks); G3, L-methionine α-deamino-γ-mercaptomethane lyase (recombinant methioninase [rMETase]) (100 U/mouse, i.p., daily, for 2 weeks). Tumor size and body weight were measured with calipers and a digital balance twice a week. The methionine level of supernatants derived from sonicated tumors was also measured. rMETase inhibited tumor growth, measured by tumor volume, compared to untreated controls and the DOX-treated group on day 14 after initiation of treatment: control (G1): 347.6 ± 88 mm3 ; DOX (G2): 329.5 ± 79 mm3 , P = 0.670; rMETase (G3): 162.6 ± 51 mm3 , P = 0.0003. The mouse body weight of the treated mice was not significantly different from the untreated controls. Tumor L-methionine levels were reduced after the rMETase-treatment compared to untreated control and pre-rMETase treatment. We previously reported efficacy of rMETase against Ewing's sarcoma and melanoma in a PDOX models. These studies suggest clinical development of rMETase, especially in recalcitrant cancers such as sarcoma.
- Published
- 2018
- Full Text
- View/download PDF
29. Simulated spaceflight-induced cardiac remodeling is modulated by gut microbial-derived trimethylamine N-oxide
- Author
-
Zizhong Liu, Gui Luo, Ruikai Du, Guanghan Kan, Xuan Han, Guohui Zhong, Wenjuan Xing, Ying Cui, Weijia Sun, Jianwei Li, Yuheng Li, Dingsheng Zhao, Xinxin Yuan, Xiaoyan Jin, Yanping Han, Huiyuan Sun, Shukuan Ling, and Yingxian Li
- Subjects
Microbial genetics ,Microbial metabolism ,Cell biology ,Metabolomics ,Model organism ,Microgravity sciences ,Science - Abstract
Summary: Spaceflight is physically demanding and can negatively affect astronauts’ health. It has been shown that the human gut microbiota and cardiac function are affected by spaceflight and simulated spaceflight. This study investigated the effects of the gut microbiota on simulated spaceflight-induced cardiac remodeling using 10° of head-down bed rest (HDBR) in rhesus macaques and 30° of hindlimb unloading (HU) in mice. The gut microbiota, fecal metabolites, and cardiac remodeling were markedly affected by HDBR in macaques and HU in mice, cardiac remodeling in control mice was affected by the gut microbiota of HU mice and that of HU mice was protected by the gut microbiota of control mice, and there was a correlation between cardiac remodeling and the gut microbial-derived metabolite trimethylamine N-oxide. These findings suggest that spaceflight can affect cardiac remodeling by modulating the gut microbiota and fecal metabolites.
- Published
- 2023
- Full Text
- View/download PDF
30. Targeting methionine with oral recombinant methioninase (o-rMETase) arrests a patient-derived orthotopic xenograft (PDOX) model of BRAF-V600E mutant melanoma: implications for chronic clinical cancer therapy and prevention
- Author
-
Masuyo Miyake, Yunfeng Li, Arun S. Singh, Sarah M. Dry, Mark A. Eckardt, Bartosz Chmielowski, Robert M. Hoffman, Kentaro Igarashi, Kentaro Miyake, Kei Kawaguchi, Yuying Tan, Shukuan Li, Michiaki Unno, Tara A. Russell, Tasuku Kiyuna, Scott D. Nelson, Fritz C. Eilber, and Qinghong Han
- Subjects
0301 basic medicine ,Nude ,Administration, Oral ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Oral administration ,Melanoma ,Cancer ,methionine dependence ,Recombinant Proteins ,nude mice ,Carbon-Sulfur Lyases ,030220 oncology & carcinogenesis ,Administration ,Female ,Proto-Oncogene Proteins B-raf ,Oral ,Mice, Nude ,Biology ,03 medical and health sciences ,PDOX ,In vivo ,Report ,medicine ,melanoma ,Animals ,Humans ,pyridoxal-L-phosphate ,Molecular Biology ,orthotopic ,Aged ,Recombinant methioninase ,Methionine ,Cancer prevention ,oral administration ,Prevention ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,In vitro ,Brain Disorders ,030104 developmental biology ,chemistry ,Cancer cell ,Mutation ,Cancer research ,Biochemistry and Cell Biology ,Developmental Biology - Abstract
The elevated methionine (MET) use by cancer cells is termed MET dependence and may be the only known general metabolic defect in cancer. Targeting MET by recombinant methioninase (rMETase) can arrest the growth of cancer cells in vitro and in vivo. We previously reported that rMETase, administrated by intra-peritoneal injection (ip-rMETase), could inhibit tumor growth in a patient-derived orthotopic xenograft (PDOX) model of a BRAF-V600E mutant melanoma. In the present study, we compared ip-rMETase and oral rMETase (o-rMETase) for efficacy on the melanoma PDOX. Melanoma PDOX nude mice were randomized into four groups of 5 mice each: untreated control; ip-rMETase (100 units, i.p., 14 consecutive days); o-rMETase (100 units, p.o., 14 consecutive days); o-rMETase+ip-rMETase (100 units, p.o.+100 units, i.p., 14 consecutive days). All treatments inhibited tumor growth on day 14 after treatment initiation, compared to untreated control (ip-rMETase, p
- Published
- 2018
31. Intra-tumor L-methionine level highly correlates with tumor size in both pancreatic cancer and melanoma patient-derived orthotopic xenograft (PDOX) nude-mouse models
- Author
-
Kentaro Igarashi, Arun S. Singh, Robert M. Hoffman, Kentaro Miyake, Scott D. Nelson, Sarah M. Dry, Yuying Tan, Mark A. Eckardt, Michiaki Unno, Masuyo Miyake, Bartosz Chemielwski, Tara A. Russell, Fritz C. Eilber, Shukuan Li, Tasuku Kiyuna, Qinghong Han, Yunfeng Li, and Kei Kawaguchi
- Subjects
0301 basic medicine ,recombinant methionine (rMETase) ,Oncology and Carcinogenesis ,tumor methionine ,pancreatic cancer ,law.invention ,Pancreatic Cancer ,03 medical and health sciences ,chemistry.chemical_compound ,Methionine level ,Rare Diseases ,0302 clinical medicine ,Nude mouse ,law ,Pancreatic cancer ,melanoma ,Medicine ,Cancer ,methionine dependence ,Methionine ,Tumor size ,biology ,business.industry ,Melanoma ,medicine.disease ,biology.organism_classification ,recombinant methionine ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Recombinant DNA ,Digestive Diseases ,business ,Research Paper - Abstract
An excessive requirement for methionine (MET) for growth, termed MET dependence, appears to be a general metabolic defect in cancer. We have previously shown that cancer-cell growth can be selectively arrested by MET restriction such as with recombinant methioninase (rMETase). In the present study, we utilized patient-derived orthotopic xenograft (PDOX) nude mouse models with pancreatic cancer or melanoma to determine the relationship between intra-tumor MET level and tumor size. After the tumors grew to 100 mm3, the PDOX nude mice were divided into two groups: untreated control and treated with rMETase (100 units, i.p., 14 consecutive days). On day 14 from initiation of treatment, intra-tumor MET levels were measured and found to highly correlate with tumor volume, both in the pancreatic cancer PDOX (p
- Published
- 2017
32. Mechanical stimulation controls osteoclast function through the regulation of Ca2+-activated Cl− channel Anoctamin 1
- Author
-
Weijia Sun, Yuheng Li, Jianwei Li, Yingjun Tan, Xinxin Yuan, Haoye Meng, Jianting Ye, Guohui Zhong, XiaoYan Jin, Zizhong Liu, Ruikai Du, Wenjuan Xing, Dingsheng Zhao, Jinping Song, Youyou Li, Junjie Pan, Yunzhang Zhao, Qi Li, Aiyuan Wang, Shukuan Ling, Rongji Dai, and Yingxian Li
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Abstract Mechanical force loading is essential for maintaining bone homeostasis, and unloading exposure can lead to bone loss. Osteoclasts are the only bone resorbing cells and play a crucial role in bone remodeling. The molecular mechanisms underlying mechanical stimulation-induced changes in osteoclast function remain to be fully elucidated. Our previous research found Ca2+-activated Cl− channel Anoctamin 1 (Ano1) was an essential regulator for osteoclast function. Here, we report that Ano1 mediates osteoclast responses to mechanical stimulation. In vitro, osteoclast activities are obviously affected by mechanical stress, which is accompanied by the changes of Ano1 levels, intracellular Cl− concentration and Ca2+ downstream signaling. Ano1 knockout or calcium binding mutants blunts the response of osteoclast to mechanical stimulation. In vivo, Ano1 knockout in osteoclast blunts loading induced osteoclast inhibition and unloading induced bone loss and. These results demonstrate that Ano1 plays an important role in mechanical stimulation induced osteoclast activity changes.
- Published
- 2023
- Full Text
- View/download PDF
33. Abstract 3258: THOR-707, a novel not-alpha IL-2, elicits durable pharmacodynamic responses in non-human primates and efficacy as single agent and in combination with anti PD-1 in multiple syngeneic mouse models
- Author
-
Lilia K. Koriazova, Hans R. Aerni, Michael J. Pena, Jasmine Nguyen, Ken Bragstad, Shukuan Li, Marcos Milla, Yelena Pavlova, Caffaro Carolina E, Ptacin Jerod, Robert W. Herman, Kristine M. San Jose, David B. Chen, Lina Ma, Ingrid B. Joseph, and Laura K. Shawver
- Subjects
0301 basic medicine ,Cancer Research ,Necrosis ,business.industry ,T cell ,Innate lymphoid cell ,T-cell receptor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Aldesleukin ,medicine ,Cancer research ,medicine.symptom ,business ,Receptor ,Alpha chain ,CD8 ,030215 immunology - Abstract
Aldesleukin is a recombinant form of IL-2 approved for metastatic melanoma and renal cell carcinoma that induced complete, durable remissions in certain patients. Yet, its use is infrequent because of vascular leak syndrome (VLS), a severe dose-limiting adverse event stemming from the engagement of the high affinity IL-2 receptor (IL-2R) alpha chain in type 2 innate lymphoid cells, eosinophils and vascular endothelial cells. THOR-707 is a site-directed, singly pegylated form of IL-2 completely lacking IL-2R alpha chain engagement yet retaining normal binding to the intermediate affinity IL-2R beta-gamma signaling complex expressed by natural killer (NK) and CD8+ T tumor-killing cells. We studied THOR-707 pharmacokinetics (PK) and pharmacodynamics (PD) in non-human primates (NHP) to evaluate peripheral biomarkers of immune cell activation (expansion of NK, CD8+ T and CD4+ regulatory T (Treg) cells and the induction of pSTAT5 and the proliferation marker Ki67). Here we show that in NHP, THOR-707 elicits the expansion of peripheral CD8+ T cells and the persistence of that response with different administration regimes. We also show that in the mouse syngeneic colon tumor model CT-26, THOR-707 induced Ki67 and the expansion of peripheral NK and CD8+ T cells. Within the tumor, THOR-707 promoted an increase in the numbers of infiltrating tumor-killing NK and CD8+ T cells without expansion of suppressive CD4+ Treg cells. Immunohistochemical (IHC) analysis showed that in THOR-707 treated animals, CD8+ T cells were mostly seen around the edges of the tumor and at areas of necrosis. Evaluation of T cell receptor (TCR) clonality revealed that treatment with THOR-707 increased intra-tumoral T cell diversity compared to the untreated animals. THOR-707 induction of CD8+ T cell tumor infiltration resulted in single agent dose-dependent anti-tumor efficacy, and additive efficacy in combination with PD-1 checkpoint inhibitor, increasing the survival of CT-26 tumor-bearing mice over either group alone. The pharmacodynamics, T cell infiltration and clonality, and efficacy of THOR-707 in two additional syngeneic models, have also been studied. Filing of an investigational new drug application for THOR-707 is expected in the second quarter of 2019 and thereafter initiation of a Phase 1/2 clinical trial in multiple tumor types as a single agent and in combination with an immune checkpoint inhibitor. Citation Format: Ingrid B. Joseph, Lina Ma, Jerod L. Ptacin, Carolina E. Caffaro, Hans R. Aerni, Kristine M. San Jose, Michael J. Pena, Robert W. Herman, Yelena Pavlova, David B. Chen, Ken Bragstad, Shukuan Li, Jasmine Nguyen, Laura K. Shawver, Lilia K. Koriazova, Marcos E. Milla. THOR-707, a novel not-alpha IL-2, elicits durable pharmacodynamic responses in non-human primates and efficacy as single agent and in combination with anti PD-1 in multiple syngeneic mouse models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3258.
- Published
- 2019
- Full Text
- View/download PDF
34. Tumor-specific cell-cycle decoy by Salmonella typhimurium A1-R combined with tumor-selective cell-cycle trap by methioninase overcome tumor intrinsic chemoresistance as visualized by FUCCI imaging
- Author
-
Toshiyoshi Fujiwara, Robert M. Hoffman, Ming Zhao, Shuya Yano, Michael Bouvet, Qinghong Han, Shukuan Li, Kiyoto Takehara, and Yuying Tan
- Subjects
0301 basic medicine ,Salmonella typhimurium ,medicine.medical_treatment ,Nude ,Drug Resistance ,Imaging ,S Phase ,Mice ,0302 clinical medicine ,cisplatinum ,Neoplasms ,Cytotoxicity ,FUCCI ,stomach cancer ,Cell Cycle ,Cell sorting ,Cell cycle ,nude mice ,Recombinant Proteins ,Carbon-Sulfur Lyases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,trap ,medicine.drug ,G2 Phase ,Paclitaxel ,Mice, Nude ,Antineoplastic Agents ,Biology ,Salmonella typhimurium A1-R ,Fluorescence ,03 medical and health sciences ,Imaging, Three-Dimensional ,medicine ,cancer ,Animals ,Humans ,Molecular Biology ,decoy ,methioninase ,Cisplatin ,Chemotherapy ,Ubiquitination ,Cancer ,Cell Biology ,medicine.disease ,030104 developmental biology ,cell-cycle ,Drug Resistance, Neoplasm ,Hela Cells ,Cancer cell ,Immunology ,Three-Dimensional ,Cancer research ,Neoplasm ,Bone marrow ,Biochemistry and Cell Biology ,Digestive Diseases ,Developmental Biology ,Reports ,HeLa Cells - Abstract
We previously reported real-time monitoring of cell cycle dynamics of cancer cells throughout a live tumor intravitally using a fluorescence ubiquitination cell cycle indicator (FUCCI). Approximately 90% of cancer cells in the center and 80% of total cells of an established tumor are in G0/G1 phase. Longitudinal real-time FUCCI imaging demonstrated that cytotoxic agents killed only proliferating cancer cells at the surface and, in contrast, and had little effect on the quiescent cancer cells. Resistant quiescent cancer cells restarted cycling after the cessation of chemotherapy. Thus cytotoxic chemotherapy which targets cells in S/G2/M, is mostly ineffective on solid tumors, but causes toxic side effects on tissues with high fractions of cycling cells, such as hair follicles, bone marrow and the intestinal lining. We have termed this phenomenon tumor intrinsic chemoresistance (TIC). We previously demonstrated that tumor-targeting Salmonella typhimurium A1-R (S. typhimurium A1-R) decoyed quiescent cancer cells in tumors to cycle from G0/G1 to S/G2/M demonstrated by FUCCI imaging. We have also previously shown that when cancer cells were treated with recombinant methioninase (rMETase), the cancer cells were selectively trapped in S/G2, shown by cell sorting as well as by FUCCI. In the present study, we show that sequential treatment of FUCCI-expressing stomach cancer MKN45 in vivo with S. typhimurium A1-R to decoy quiescent cancer cells to cycle, with subsequent rMETase to selectively trap the decoyed cancer cells in S/G2 phase, followed by cisplatinum (CDDP) or paclitaxel (PTX) chemotherapy to kill the decoyed and trapped cancer cells completely prevented or regressed tumor growth. These results demonstrate the effectiveness of the praradigm of "decoy, trap and shoot" chemotherapy.
- Published
- 2016
35. Abstract P4-07-04: Methioninase cell-cycle synchronization potentiates chemotherapy for breast cancer
- Author
-
S Yano, Qinghong Han, Shukuan Li, RM Hoffman, Yuying Tan, and Toshiyoshi Fujiwara
- Subjects
Cancer Research ,education.field_of_study ,business.industry ,Cell ,Population ,Cancer ,Cell cycle ,medicine.disease ,medicine.anatomical_structure ,Breast cancer ,Oncology ,Cancer cell ,Immunology ,medicine ,Cancer research ,Doxorubicin ,business ,Cell synchronization ,education ,medicine.drug - Abstract
Deprivation of methionine selectively arrests cancer cells during late S-phase (Proc. Natl. Acad. Sci. USA 77, 7306–7310, 1980; Biochim. Biophys. Acta, Reviews on Cancer 738, 49–87, 1984), where they are highly sensitive to chemotherapy drugs which damage DNA (J. Natl. Cancer Inst. 76, 629–639, 1986). Cancer cells, transformed to express different color fluorescent reporters during specific phases of the cell cycle (Cell 132, 487–498, 2008), were used to monitor the onset of the S/G2-phase block due to methionine deprivation effected by recombinant methioninase (rMETase). The S/G2-phase blocked cancer cells fluoresced yellow or green in contrast to cancer cells in G1 which fluoresced red. Cancer cells, including MCF-7 breast cancer, synchronously blocked in S/G2-phase by rMETase, were identified by their yellow-green fluorescence and allowed to accumulate to the maximum extent. At the point of maximum yellow/green cells in the culture, the cells were administered chemotherapy drugs which interact with DNA or block DNA synthesis such as doxorubicin. We termed this procedure color-coded chemotherapy (CCC). CCC was highly effective against the cancer cells (90% cell kill). In contrast, treatment of cancer cells with drugs only, and without rMETase-effected S/G2-phase synchrony, led to the majority of the cancer cell population being blocked in G1 phase (red fluorescent) where they were resistant to the drugs (40% cell kill). CCC, which identifies, by fluorescent color, when cancer cells are blocked in S/G2-phase by a unique cell-cycle-blocking agent, rMETase, demonstrates the potential of cell-synchronization-based chemotherapy for breast cancer. Citation Information: Cancer Res 2012;72(24 Suppl):Abstract nr P4-07-04.
- Published
- 2012
- Full Text
- View/download PDF
36. PEGylation Confers Greatly Extended Half-Life and Attenuated Immunogenicity to Recombinant Methioninase in Primates
- Author
-
Akio Takimoto, Junhua Wang, Eugene P. Frenkel, Jinbao Xu, Takayuji Yoshioka, Yuying Tan, Dongdong Zhang, Robert M. Hoffman, Xinghua Sun, Quan Lu, Shigeo Yagi, Shukuan Li, Zhijian Yang, Tomoaki Takakura, Yoshinao Kobayashi, Changgen Lian, Ying Zhang, and Chunmei Chen
- Subjects
Male ,Cancer Research ,Antigenicity ,Biology ,Pharmacology ,Antibodies ,Polyethylene Glycols ,Eating ,chemistry.chemical_compound ,Methionine ,Pharmacokinetics ,Animals ,Drug Carriers ,Dose-Response Relationship, Drug ,Body Weight ,Half-life ,Recombinant Proteins ,Carbon-Sulfur Lyases ,Macaca fascicularis ,Dose–response relationship ,Oncology ,chemistry ,Toxicity ,Immunology ,biology.protein ,PEGylation ,Antibody ,Half-Life - Abstract
Methionine depletion by recombinant methioninase (rMETase) has been demonstrated previously to be highly effective in tumor-bearing mouse models. However, the therapeutic potential of rMETase has been limited by its short plasma half-life and immunologic effects, including high antibody production in mice and monkeys and anaphylactic reactions in monkeys. To overcome these limits of rMETase, the enzyme has been coupled to methoxypolyethylene glycol succinimidyl glutarate (MEGC-PEG-5000). In this study, we evaluated the pharmacokinetics, antigenicity and toxicity of MEGC-PEG-rMETase in Macaca fascicularis monkeys using an escalating-dose strategy. Dose ranging studies at 1,000, 4,000, and 8,000 units/kg i.v. determined that a single dose of 4,000 units/kg was sufficient to reduce plasma methionine to
- Published
- 2004
- Full Text
- View/download PDF
37. Circulating Half-Life of PEGylated Recombinant Methioninase Holoenzyme Is Highly Dose Dependent on Cofactor Pyridoxal-5′-Phosphate
- Author
-
Yoshinao Kobayashi, Xiaoen Wang, Robert M. Hoffman, Xinghua Sun, Yuying Tan, Shigeo Yagi, Zhijian Yang, Shukuan Li, Takayuki Yoshioka, Tomoaki Takakura, Nan Zhang, Akinori Suginaka, Akio Takimoto, and Eugene P. Frenkel
- Subjects
Antimetabolites, Antineoplastic ,Cancer Research ,Mice, Nude ,chemical and pharmacologic phenomena ,Pharmacology ,Cofactor ,Polyethylene Glycols ,law.invention ,Mice ,chemistry.chemical_compound ,immune system diseases ,In vivo ,law ,PEG ratio ,Animals ,chemistry.chemical_classification ,Mice, Inbred BALB C ,Methionine ,Dose-Response Relationship, Drug ,biology ,technology, industry, and agriculture ,Half-life ,Recombinant Proteins ,nervous system diseases ,Carbon-Sulfur Lyases ,Dose–response relationship ,Enzyme ,Oncology ,chemistry ,Biochemistry ,Pyridoxal Phosphate ,biology.protein ,Recombinant DNA ,lipids (amino acids, peptides, and proteins) ,Half-Life - Abstract
Recombinant methioninase (rMETase) has been shown to target the elevated methionine (MET) dependence of tumor cells and arrest their growth as well as make tumors more sensitive to standard chemotherapy agents. Polyethylene glycol (PEG)-modified rMETase (PEG-rMETase) has reduced antigenicity compared with unmodified rMETase. However, PEG-rMETase has a limited active circulating half-life due to rapid in vivo dissociation of its cofactor pyridoxal-5′-phosphate (PLP), a surprising finding, because PLP is tightly bound to PEG-rMETase in buffer. The question asked in the current study was on the effect of increasing doses of PLP to extend the circulating half-life of active PEG-rMETase holoenzyme in vivo. rMETase was conjugated with methoxypolyethylene glycol succinimidyl glutarate 5000 (MEGC-PEG). Miniosmotic pumps containing various concentrations of PLP were implanted in BALB-C mice. PLP-infused mice were then injected with a single dose of 4000 or 8000 units/kg PEG-rMETase. Mice infused with 5, 50, 100, 200, and 500 mg/ml PLP-containing miniosmotic pumps increased plasma PLP to 7, 24, 34, 60, and 95 μm, respectively, from the PLP baseline of 0.3 μm. PLP increased the half-life of MEGC-PEG-rMETase holoenzyme in a dose-dependent manner. Pumps containing 500 mg/ml PLP increased the half-life of MEGC-PEG-rMETase holoenzyme 4.5-fold from 1.5 to 7 h. Infused PLP did not extend the half-life of MEGC-PEG-rMETase apoenzyme. With a dose of 4000 units/kg MEGC-PEG-rMETase in the mice infused with 5, 50, 200, and 500 mg/ml PLP, plasma MET was depleted from 50 μm to ≤5 μm for 8, 24, 72, and 72 h, respectively. Thus, PLP infusion could extend the period of MET depletion by MEGC-PEG-rMETase by ∼10-fold in a dose-dependent manner. The mice given 8000 units/kg MEGC-PEG-rMETase showed a similar plasma MET depletion time course, indicating that the limiting factor for MEGC-PEG-rMETase-mediated MET depletion in vivo was PLP. The extended time of MET depletion by MEGC-PEG-rMETase was due to the maintenance of active MEGC-PEG-rMETase holoenzyme by infused PLP. The infused PLP either bound to apo-MEGC-PEG-rMETase and/or inhibited dissociation of PLP from holo-PEG-rMETase, thereby maintaining the holoenzyme form of MEGC-PEG-rMETase in vivo. The combination of MEGC-PEG-rMETase treatment with PLP infusion suggests an effective clinical strategy for long-term MET depletion to arrest cancer growth.
- Published
- 2004
- Full Text
- View/download PDF
38. Pharmacokinetics, Methionine Depletion, and Antigenicity of Recombinant Methioninase in Primates
- Author
-
Eugene P. Frenkel, Takayuki Yoshioka, Quan Lu, Robert M. Hoffman, Junhua Wang, Xinghua Sun, Shukuan Li, Shigeo Yagi, Zhijian Yang, Yuying Tan, and Baoqiu Li
- Subjects
Male ,Primates ,Cancer Research ,medicine.medical_specialty ,Antigenicity ,Serum albumin ,Biology ,chemistry.chemical_compound ,Methionine ,Pharmacokinetics ,Internal medicine ,medicine ,Animals ,Humans ,Neutralizing antibody ,Recombinant Proteins ,Carbon-Sulfur Lyases ,Kinetics ,Macaca fascicularis ,Endocrinology ,Oncology ,chemistry ,Toxicity ,biology.protein ,Antibody ,Half-Life ,Blood sampling - Abstract
Pharmacokinetics, methionine depletion, antigenicity, and toxicity of recombinant methioninase (rMETase), which has shown efficacy in achieving cell kill in a broad range of human tumor models, were examined in macaque monkeys. Dose-ranging studies at 1000, 2000, and 4000 units/kg i.v. identified the 4000 units/kg dose as able to reduce plasma methionine to an undetectable level (less than 0.5 μm) by 30 min, and the level so remained for 8 h. Pharmacokinetic analysis showed that rMETase was eliminated with a T1/2 of 2.49 h. A 2-week i.v. administration of 4000 units/kg every 8 h/day for 2 weeks resulted in a steady-state depletion of plasma methionine to less than 2 μm. The only manifest toxicity was decreased food intake and slight weight loss. Serum albumin and red cell values declined transiently during treatment, which may be related to extensive blood sampling. Re-challenge on day 28 resulted in anaphylactic shock and death in one animal. Subsequent pretreatment with hydrocortisone prevented the anaphylactic reaction, although vomiting was frequently observed. Re-challenge was carried out at days 66, 86, and 116. Anti-rMETase antibodies (at 10−3) were found after the first challenge, and these increased to 10−6 after the fourth challenge and decreased to 10−2 by 2 months post therapy. The main rMETase antibody was IgG, and although it has some in vitro features of being a neutralizing antibody, each challenge dose was effective in depleting plasma methionine levels. Thus, rMETase was able to effectively deplete plasma methionine levels with minimal toxicity in a primate model. These data provide the bases for alteration by polyethyleneglycol conjugation (PEGylation) of the enzyme to increase its duration of effect and reduce its immunogenicity.
- Published
- 2004
- Full Text
- View/download PDF
39. Anoctamin 1 controls bone resorption by coupling Cl− channel activation with RANKL-RANK signaling transduction
- Author
-
Weijia Sun, Shuai Guo, Yuheng Li, JianWei Li, Caizhi Liu, Yafei Chen, Xuzhao Wang, Yingjun Tan, Hua Tian, Cheng Wang, Ruikai Du, Guohui Zhong, Sai Shi, Biao Ma, Chang Qu, Jingxuan Fu, Xiaoyan Jin, Dingsheng Zhao, Yong Zhan, Shukuan Ling, Hailong An, and Yingxian Li
- Subjects
Science - Abstract
Osteoclast over-activation leads to bone loss and chloride homeostasis is important for osteoclast function. Here, the authors show that Anoctamin 1 controls bone resorption by coupling Cl− channel activation with RANKL-RANK signaling transduction.
- Published
- 2022
- Full Text
- View/download PDF
40. Effects of 60 days of 6° head-down bed rest on the composition and function of the human gut microbiota
- Author
-
Yixuan Li, Zizhong Liu, Gui Luo, Haiyun Lan, Pu Chen, Ruikai Du, Gongchao Jing, Lu Liu, Xiaohan Cui, Yongzhi Li, Yanping Han, Jian Xu, Hongwei Zhu, Shukuan Ling, and Yingxian Li
- Subjects
Genomics ,Genomic analysis ,Space sciences ,Science - Abstract
Summary: Spaceflight is rigorous and dangerous environment which can negatively affect astronauts’ health and the entire mission. The 60 days of 6° head-down bed rest (HDBR) experiment provided us with an opportunity to trace the change of gut microbiota under simulated microgravity. The gut microbiota of volunteers was analyzed and characterized by 16S rRNA gene sequencing and metagenomic sequencing. Our results showed that the composition and function of the volunteers’ gut microbiota were markedly was affected by 60 days of 6° HDBR. We further confirmed the species and diversity fluctuations. Resistance and virulence genes in the gut microbiota were also affected by 60 days of 6° HDBR, but the species attributions remained stable. The human gut microbiota affected by 60 days of 6° HDBR which was partially consistent with the effect of spaceflight, this implied that HDBR was a simulation of how spaceflight affects the human gut microbiota.
- Published
- 2023
- Full Text
- View/download PDF
41. Selective methioninase-induced trap of cancer cells in S/G2 phase visualized by FUCCI imaging confers chemosensitivity
- Author
-
Shukuan Li, Shuya Yano, Toshiyoshi Fujiwara, Qinghong Han, Robert M. Hoffman, Michael Bouvet, and Yuying Tan
- Subjects
Time Factors ,Antimetabolites ,medicine.medical_treatment ,Drug Resistance ,Uterine Cervical Neoplasms ,chemotherapy ,Genes, Reporter ,Antineoplastic Combined Chemotherapy Protocols ,Neoplasm ,MCF-7 cells ,Cancer ,education.field_of_study ,Microscopy ,FUCCI ,Microscopy, Confocal ,imaging ,recombinant methioninase ,Transfection ,Cell cycle ,Antineoplastic ,Recombinant Proteins ,3. Good health ,Carbon-Sulfur Lyases ,S/G(2) phase block ,Oncology ,Confocal ,S Phase Cell Cycle Checkpoints ,Female ,cell cycle ,medicine.drug ,Research Paper ,Antimetabolites, Antineoplastic ,Population ,Oncology and Carcinogenesis ,Breast Neoplasms ,Biology ,Fluorescence ,medicine ,Humans ,Doxorubicin ,HeLa cells ,education ,Reporter ,Cisplatin ,Chemotherapy ,S/G2 phase block ,medicine.disease ,Molecular biology ,rMETase ,Microscopy, Fluorescence ,Genes ,Drug Resistance, Neoplasm ,Cancer cell - Abstract
A major impediment to the response of tumors to chemotherapy is that the large majority of cancer cells within a tumor are quiescent in G0/G1, where cancer cells are resistant to chemotherapy. To attempt to solve this problem of quiescent cells in a tumor, cancer cells were treated with recombinant methioninase (rMETase), which selectively traps cancer cells in S/G2. The cell cycle phase of the cancer cells was visualized with the fluorescence ubiquitination-based cell cycle indicator cell cycle indicator (FUCCI). At the time of rMETase-induced S/G2-phase blockage, identified by the cancer cells' green fluorescence by FUCCI imaging, the cancer cells were administered S/G2-dependent chemotherapy drugs, which interact with DNA or block DNA synthesis such as doxorubicin, cisplatin, or 5-fluorouracil. Treatment of cancer cells with drugs only, without rMETase-induced S/G2 phase blockage, led to the majority of the cancer-cell population being blocked in G0/G1 phase, identified by the cancer cells becoming red fluorescent in the FUCCI system. The G0/G1 blocked cells were resistant to the chemotherapy. In contrast, trapping of cancer cells in S/G2 phase by rMETase treatment followed by FUCCI-imaging-guided chemotherapy was highly effective in killing the cancer cells.
- Published
- 2014
42. The mechanosensitive lncRNA Neat1 promotes osteoblast function through paraspeckle-dependent Smurf1 mRNA retention
- Author
-
Caizhi Liu, Xingcheng Gao, Yuheng Li, Weijia Sun, Youjia Xu, Yingjun Tan, Ruikai Du, Guohui Zhong, Dingsheng Zhao, Zizhong Liu, Xiaoyan Jin, Yinlong Zhao, Yinbo Wang, Xinxin Yuan, Junjie Pan, Guodong Yuan, Youyou Li, Wenjuan Xing, Guanghan Kan, Yanqing Wang, Qi Li, Xuan Han, Jianwei Li, Shukuan Ling, and Yingxian Li
- Subjects
Biology (General) ,QH301-705.5 ,Physiology ,QP1-981 - Abstract
Abstract Mechanical stimulation plays an important role in bone remodeling. Exercise-induced mechanical loading enhances bone strength, whereas mechanical unloading leads to bone loss. Increasing evidence has demonstrated that long noncoding RNAs (lncRNAs) play key roles in diverse biological, physiological and pathological contexts. However, the roles of lncRNAs in mechanotransduction and their relationships with bone formation remain unknown. In this study, we screened mechanosensing lncRNAs in osteoblasts and identified Neat1, the most clearly decreased lncRNA under simulated microgravity. Of note, not only Neat1 expression but also the specific paraspeckle structure formed by Neat1 was sensitive to different mechanical stimulations, which were closely associated with osteoblast function. Paraspeckles exhibited small punctate aggregates under simulated microgravity and elongated prolate or larger irregular structures under mechanical loading. Neat1 knockout mice displayed disrupted bone formation, impaired bone structure and strength, and reduced bone mass. Neat1 deficiency in osteoblasts reduced the response of osteoblasts to mechanical stimulation. In vivo, Neat1 knockout in mice weakened the bone phenotypes in response to mechanical loading and hindlimb unloading stimulation. Mechanistically, paraspeckles promoted nuclear retention of E3 ubiquitin ligase Smurf1 mRNA and downregulation of their translation, thus inhibiting ubiquitination-mediated degradation of the osteoblast master transcription factor Runx2, a Smurf1 target. Our study revealed that Neat1 plays an essential role in osteoblast function under mechanical stimulation, which provides a paradigm for the function of the lncRNA-assembled structure in response to mechanical stimulation and offers a therapeutic strategy for long-term spaceflight- or bedrest-induced bone loss and age-related osteoporosis.
- Published
- 2022
- Full Text
- View/download PDF
43. Hydrogen Inhalation Ameliorates Oxidative Stress and Glucose Metabolism Disorder in the Brain of Hindlimb Unloading Rats
- Author
-
Xiaoyan Jin, Fei Xie, Yang Yi, Yating Zhang, Shukuan Ling, Qianwei Shen, Xuemei Ma, and Yingxian Li
- Subjects
Motor vehicles. Aeronautics. Astronautics ,TL1-4050 ,Astronomy ,QB1-991 - Abstract
Many studies have shown that spaceflight causes oxidative stress and induces brain disorder in astronauts, but the counter measurements are lacking. Increasing evidence demonstrated that hydrogen can act as a therapeutic antioxidant. In this study, rats were treated with or without about 5% hydrogen under hindlimb unloading or normal conditions for 28 d. We assessed rat’s brain function by open-field test, step-down passive avoidance test, the neurotransmitter’s level detected by liquid chromatography with tandem mass spectrometry, and Nissl and hematoxylin–eosin staining analysis. We also assessed the oxidative damage by changes of malondialdehyde level, the ratio of reduced glutathione to oxidized glutathione, and superoxide dismutase and catalase activity. Glucose metabolism disorder was disclosed through glucose metabolomic analysis. The underlying mechanism of the effects of hydrogen was analyzed by mRNA sequencing and detecting mRNA and protein levels. Our data showed that hindlimb unloading caused oxidative damage and glucose metabolism disorder in brain tissues and decreased brain function in rats. Hydrogen inhalation reduced oxidative damage, ameliorated glucose metabolism disorder, and alleviated the dysfunction of rat brain function. Peroxisome-proliferator-activated receptor gamma coactivator 1α and brain-derived neurotrophic factor, the key regulators of glucose metabolism and brain function, were obviously affected. This research confirms the protective effect of hydrogen inhalation on declining brain function under hindlimb unloading conditions and discloses the underlying mechanism, which provides a potential strategy for astronauts’ health.
- Published
- 2023
- Full Text
- View/download PDF
44. Diagnosis of Alzheimer’s Disease Based on Accelerated Mirror Descent Optimization and a Three-Dimensional Aggregated Residual Network
- Author
-
Yue Tu, Shukuan Lin, Jianzhong Qiao, Peng Zhang, and Kuankuan Hao
- Subjects
Alzheimer’s disease ,optimization algorithm ,mirror descent ,CNN ,MRI ,Chemical technology ,TP1-1185 - Abstract
Alzheimer’s disease (AD), a neuropsychiatric disorder, continually arises in the elderly. To date, no targeted medications have been developed for AD. Early and fast diagnosis of AD plays a pivotal role in identifying potential AD patients, enabling timely medical interventions, and mitigating disease progression. Computer-aided diagnosis (CAD) becomes possible with the burgeoning of deep learning. However, the existing CAD models for processing 3D Alzheimer’s disease images usually have the problems of slow convergence, disappearance of gradient, and falling into local optimum. This makes the training of 3D diagnosis models need a lot of time, and the accuracy is often poor. In this paper, a novel 3D aggregated residual network with accelerated mirror descent optimization is proposed for diagnosing AD. First, a novel unbiased subgradient accelerated mirror descent (SAMD) optimization algorithm is proposed to speed up diagnosis network training. By optimizing the nonlinear projection process, our proposed algorithm can avoid the occurrence of the local optimum in the non-Euclidean distance metric. The most notable aspect is that, to the best of our knowledge, this is the pioneering attempt to optimize the AD diagnosis training process by improving the optimization algorithm. Then, we provide a rigorous proof of the SAMD’s convergence, and the convergence of SAMD is better than any existing gradient descent algorithms. Finally, we use our proposed SAMD algorithm to train our proposed 3D aggregated residual network architecture (ARCNN). We employed the ADNI dataset to train ARCNN diagnostic models separately for the AD vs. NC task and the sMCI vs. pMCI task, followed by testing to evaluate the disease diagnostic outcomes. The results reveal that the accuracy can be improved in diagnosing AD, and the training speed can be accelerated. Our proposed method achieves 95.4% accuracy in AD diagnosis and 79.9% accuracy in MCI diagnosis; the best results contrasted with several state-of-the-art diagnosis methods. In addition, our proposed SAMD algorithm can save about 19% of the convergence time on average in the AD diagnosis model compared with the gradient descent algorithms, which is very momentous in clinic.
- Published
- 2023
- Full Text
- View/download PDF
45. Human tumor growth in nude mice is associated with decreased plasma cysteine and homocysteine
- Author
-
Farida, Al-Awadi, Meng, Yang, Yuying, Tan, Qinghong, Han, Shukuan, Li, and Robert M, Hoffman
- Subjects
Male ,Mice, Nude ,Prostatic Neoplasms ,Breast Neoplasms ,Cell Growth Processes ,Pancreatic Neoplasms ,Mice ,Methionine ,Cell Line, Tumor ,Neoplasms ,Colonic Neoplasms ,Biomarkers, Tumor ,Animals ,Humans ,Female ,Cysteine ,Homocysteine ,Neoplasm Transplantation - Abstract
The methionine cycle and its metabolites homocysteine and cysteine serve several important functions in cellular metabolism. Abnormalities in metabolism of the methionine cycle have been associated with cancer. We determined plasma levels of methionine, homocysteine and cysteine in nude mice implanted with human cancer cell lines (MDA-MB-435 breast, PC-3 prostate, HT29 colon, BX-PC3 pancreas) over a prolonged period of tumor growth. The data were compared with correspondins values in nontumor-bearing controls. Nude mice were injected s.c. in the right flank with 10(6) cancer cells. Tumor growth was measured over time. Methionine was measured in plasma by HPLC. Cysteine and homocysteine were measured in plasma by recombinant enzyme assays and spectrophotometry to measure products. The concentrations of cysteine and homocysteine in plasma decreased significantly as a result of progression of breast, prostate and the pancreas tumor types implanted in the nude mice at least over a two-month period. Data for the colon tumors were nonsignificant for both cysteine and homocysteine. In the case of methionine, the decrease was significant only due to progression of the breast tumors, grown over a long time period, as compared to the mice without tumors control. The results suggest that sulphur amino acids may be plasma or serum biomarkers for cancer progression.
- Published
- 2008
46. Effect of methionine-depletion via methioninase-treatment on cancer cells in S/G2 phase and chemosensitivity
- Author
-
Yuying Tan, Robert M. Hoffman, Shuya Yano, Toshiyoshi Fujiwara, Michael Bouvet, Qinghong Han, and Shukuan Li
- Subjects
Cancer Research ,chemistry.chemical_compound ,Chemotherapy ,Methionine ,Oncology ,chemistry ,business.industry ,medicine.medical_treatment ,Cancer cell ,Cancer research ,Medicine ,business - Abstract
e13512 Background: A major impediment to the response of tumors to chemotherapy is that the large majority of cancer cells within a tumor are quiescent in G0/G1, where cancer cells are resistant to...
- Published
- 2015
- Full Text
- View/download PDF
47. Non-Invasive Skin Imaging Assessment of Human Stress During Head-Down Bed Rest Using a Portable Handheld Two-Photon Microscope
- Author
-
Junjie Wang, Zhen Zhen, Yanqing Wang, Runlong Wu, Yanhui Hu, Qiang Fu, Yongzhi Li, Bingmu Xin, Jinping Song, Jianwei Li, Yafei Ren, Lishuang Feng, Heping Cheng, Aimin Wang, Liming Hu, Shukuan Ling, and Yingxian Li
- Subjects
skin ,portable handheld two-photon microscope ,TPEF ,SHG ,head-down bed rest ,Physiology ,QP1-981 - Abstract
Spaceflight presents a series of physiological and pathological challenges to astronauts resulting from ionizing radiation, microgravity, isolation, and other spaceflight hazards. These risks cause a series of aging-related diseases associated with increased oxidative stress and mitochondria dysfunction. The skin contains many autofluorescent substances, such as nicotinamide adenine dinucleotide phosphate (NAD(P)H), keratin, melanin, elastin, and collagen, which reflect physiological and pathological changes in vivo. In this study, we used a portable handheld two-photon microscope to conduct high-resolution in vivo skin imaging on volunteers during 15 days of head-down bed rest. The two-photon microscope, equipped with a flexible handheld scanning head, was used to measure two-photon excited fluorescence (TPEF) and second harmonic generation (SHG) images of the left forearm, left front chest, and forehead of volunteers. Changes in TPEF, SHG, and the extended SHG-to-AF(TPEF) aging index of the dermis (SAAID) were measured. It was found that TPEF intensity increased during bed rest and was restored to normal levels after recovery. Meanwhile, SHG increased slightly during bed rest, and the skin aging index increased. Moreover, we found the skin TPEF signals of the left forearm were significantly negatively associated with the oxidative stress marker malondialdehyde (MDA) and DNA damage marker 8-hydroxy-2′-desoxyguanosine (8-OHdG) values of subjects during head-down bed rest. Meanwhile, the SHG signals were also significantly negatively correlated with MDA and 8-OHDG. A significant negative correlation between the extended SAAID of the left chest and serum antioxidant superoxide dismutase (SOD) levels was also found. These results demonstrate that skin autofluorescence signals can reflect changes in human oxidant status. This study provides evidence for in-orbit monitoring of changes in human stress using a portable handheld two-photon microscope for skin imaging.
- Published
- 2022
- Full Text
- View/download PDF
48. A rapid HPLC method for the measurement of ultra-low plasma methionine concentrations applicable to methionine depletion therapy
- Author
-
Xinghua, Sun, Yuying, Tan, Zhijian, Yang, Shukuan, Li, and Robert M, Hoffman
- Subjects
Mice ,Methionine ,Sodium Acetate ,Methanol ,Calibration ,Linear Models ,Animals ,Furans ,Sensitivity and Specificity ,Chromatography, High Pressure Liquid - Abstract
A rapid, sensitive and specific high-performance liquid chromatographic (HPLC) method for the separation and quantification of L-methionine in plasma has been developed. After derivatization of plasma amino acids with o-phthalaldehyde (OPA), a 50 microl sample was loaded on a reversed-phase Supelcosil LC-18-DB column (particle size 5 microm, 25 cm x 4.6 mm, 120A pores). A customized gradient program using tetrahydrofuran/methanol/0.1 M sodium acetate, pH 7.0, v/v/v=5/95/900 and methanol was used with detection by fluorescence. The elution time was 15 minutes, a 3-fold improvement over existing methods. The linearity was 1-100 microM. The limit of detection was 0.5 micromol/L, a 10-fold improvement over existing methods. The inter-assay CVs were 2-5%, and the intra-assays CVs were 4-8%. The sensitivity and rapidity of this HPLC method is particularly applicable to determine the efficacy of methionine depletion therapy of cancer.
- Published
- 2005
49. Tumor-targeting bacterial therapy with amino acid auxotrophs of GFP-expressing Salmonella typhimurium
- Author
-
Ming Zhao, Sheldon Penman, Meng Yang, Eugene Baranov, Shukuan Li, Ping Jiang, Mingxu Xu, Xiao-Ming Li, and Robert M. Hoffman
- Subjects
Male ,Salmonella typhimurium ,Green Fluorescent Proteins ,Mice, Nude ,Genetically modified bacteria ,Spleen ,Green fluorescent protein ,Microbiology ,Mice ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Amino Acids ,Selection, Genetic ,Multidisciplinary ,biology ,Bacteria ,Cell Death ,Prostatic Neoplasms ,Neoplasms, Experimental ,Biological Sciences ,biology.organism_classification ,In vitro ,Biological Therapy ,medicine.anatomical_structure ,Treatment Outcome ,Cell culture ,Cancer cell ,Neoplasm Transplantation - Abstract
Here we report a genetically modified bacteria strain, Salmonella typhimurium A1, selected for anticancer activity in vivo . The strain grows in tumor xenografts. In sharp contrast, normal tissue is cleared of these bacteria even in immunodeficient athymic mice. S . typhimurium A1 is auxotrophic (Leu/Arg-dependent) but apparently receives sufficient support from the neoplastic tissue to grow locally. Whether additional genetic lesions are present is not known. In in vitro infection, the GFP-expressing bacteria grew in the cytoplasm of PC-3 human prostate cancer cells and caused nuclear destruction. These effects were visualized in cells labeled with GFP in the nucleus and red fluorescent protein in the cytoplasm. In vivo , the bacteria caused tumor inhibition and regression of xenografts visualized by whole-body imaging. The bacteria, introduced i.v. or intratumorally, invaded and replicated intracellularly in PC-3 prostate cancer cells labeled with red fluorescent protein grafted into nude mice. By day 15, S . typhimurium A1 was undetectable in the liver, lung, spleen, and kidney, but it continued to proliferate in the PC-3 tumor, which stopped growing. When the bacteria were injected intratumorally, the tumor completely regressed by day 20. There were no obvious adverse effects on the host when the bacteria were injected by either route. The S . typhimurium A1 strain grew throughout the tumor, including viable malignant tissue. This result is in marked contrast to bacteria previously tried for cancer therapy that were confined to necrotic areas of the tumor, which may account, in part, for the strain's unique antitumor efficacy.
- Published
- 2005
50. Protein carboxyl amidation increases the potential extent of protein polyethylene glycol conjugation
- Author
-
Robert M. Hoffman, Shukuan Li, Xinghua Sun, Shigeo Yagi, Yuying Tan, and Zhijian Yang
- Subjects
chemistry.chemical_classification ,Binding Sites ,Protein subunit ,technology, industry, and agriculture ,Biophysics ,Proteins ,Succinimides ,Cell Biology ,Polyethylene glycol ,Biochemistry ,Amides ,law.invention ,Polyethylene Glycols ,chemistry.chemical_compound ,Enzyme ,chemistry ,law ,PEG ratio ,PEGylation ,Recombinant DNA ,Putrescine ,Binding site ,Molecular Biology ,Carbodiimide - Abstract
Chemical coupling of polyethylene glycol (PEG) to therapeutic proteins reduces their immunogenicity and prolongs their circulating half-life. The limitation of this approach is the number and distribution of sites on proteins available for PEGylation (the N terminus and the -amino group of lysines). To increase the extent of PEGylation, we have developed a method to increase the number of PEGylation sites in a model protein, recombinant methionine alpha,gamma-lyase (recombinant methioninase; rMETase), an enzyme cancer therapeutic cloned from Pseudomonas putida. rMETase was first PEGylated with methoxypolyethylene glycol succinimidyl glutarate-5000 with a molar ratio of PEG:rMETase of 15:1. The carboxyl groups of the initially PEGylated protein were then conjugated with diaminobutane, resulting in carboxyl amidation. This reaction was catalyzed by 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide, a water-soluble carbodiimide. The steric hindrance provided by the PEG chains already coupled to the protein prevented cross-linking between rMETase molecules during the carboxyl amidation reaction. The carboxyl-amidated PEGylated rMETase was hyper-PEGylated at a molar ratio of PEG to PEG-rMETase of 60:1. Biochemical analysis indicated that 13 PEG chains were coupled to each subunit of rMETase after hyper-PEGylation compared with 6-8 PEG chains attached to the non-carboxyl-amidated PEG-rMETase. Approximately 15-20% of the non-PEGylated rMETase activity was retained in the hyper-PEGylated molecule. Immunogenicity of the hyper-PEG-rMETase was significantly reduced relative to PEG-rMETase and rMETase. Initial results suggest that hyper-PEGylation may become a new strategy for PEGylation of protein biologics.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.