157 results on '"Khalid S. Mohammad"'
Search Results
2. Intersecting Paths: Unraveling the Complex Journey of Cancer to Bone Metastasis
- Author
-
Nour Arakil, Shahid Akhtar Akhund, Basant Elaasser, and Khalid S. Mohammad
- Subjects
bone metastases ,bone microenvironment ,bone remodeling ,interleukin ,therapy ,Biology (General) ,QH301-705.5 - Abstract
The phenomenon of bone metastases presents a significant challenge within the context of advanced cancer treatments, particularly pertaining to breast, prostate, and lung cancers. These metastatic occurrences stem from the dissemination of cancerous cells into the bone, thereby interrupting the equilibrium between osteoblasts and osteoclasts. Such disruption results in skeletal complications, adversely affecting patient morbidity and quality of life. This review discusses the intricate interplay between cancer cells and the bone microenvironment, positing the bone not merely as a passive recipient of metastatic cells but as an active contributor to cancer progression through its distinctive biochemical and cellular makeup. A thorough examination of bone structure and the dynamics of bone remodeling is undertaken, elucidating how metastatic cancer cells exploit these processes. This review explores the genetic and molecular pathways that underpin the onset and development of bone metastases. Particular emphasis is placed on the roles of cytokines and growth factors in facilitating osteoclastogenesis and influencing osteoblast activity. Additionally, this paper offers a meticulous critique of current diagnostic methodologies, ranging from conventional radiography to advanced molecular imaging techniques, and discusses the implications of a nuanced understanding of bone metastasis biology for therapeutic intervention. This includes the development of targeted therapies and strategies for managing bone pain and other skeletal-related events. Moreover, this review underscores the imperative of ongoing research efforts aimed at identifying novel therapeutic targets and refining management approaches for bone metastases. It advocates for a multidisciplinary strategy that integrates advancements in medical oncology and radiology with insights derived from molecular biology and genetics, to enhance prognostic outcomes and the quality of life for patients afflicted by this debilitating condition. In summary, bone metastases constitute a complex issue that demands a comprehensive and informed approach to treatment. This article contributes to the ongoing discourse by consolidating existing knowledge and identifying avenues for future investigation, with the overarching objective of ameliorating patient care in the domain of oncology.
- Published
- 2024
- Full Text
- View/download PDF
3. Aging‐associated skeletal muscle defects in HER2/Neu transgenic mammary tumour model
- Author
-
Ruizhong Wang, Brijesh Kumar, Poornima Bhat‐Nakshatri, Mayuri S. Prasad, Max H. Jacobsen, Gabriela Ovalle, Calli Maguire, George Sandusky, Trupti Trivedi, Khalid S. Mohammad, Theresa Guise, Narsimha R. Penthala, Peter A. Crooks, Jianguo Liu, Teresa Zimmers, and Harikrishna Nakshatri
- Subjects
Breast cancer ,Functional limitations ,Skeletal muscle ,Cytokines/chemokines ,NF‐κB ,Internal medicine ,RC31-1245 - Abstract
Abstract Background Loss of skeletal muscle volume and functional limitations are poor prognostic markers in breast cancer patients. Several molecular defects in skeletal muscle including reduced myoblast determination protein 1 (MyoD) levels and increased protein turn over due to enhanced proteosomal activity have been suggested as causes of skeletal muscle loss in cancer patients. However, it is unknown whether molecular defects in skeletal muscle are dependent on tumour aetiology. Methods We characterized functional and molecular defects of skeletal muscle in mouse mammary tumour virus (MMTV)‐Neu (Neu+) mice (n = 6–12), an animal model that represents HER2 + human breast cancer, and compared the results with well‐characterized luminal B breast cancer model MMTV‐PyMT (PyMT+). Functional studies such as grip strength, rotarod performance, and ex vivo muscle contraction were performed to measure the effects of cancer on skeletal muscle. Expression of muscle‐enriched genes and microRNAs as well as circulating cytokines/chemokines were measured. Because nuclear factor‐kappaB (NF‐κB) pathway plays a significant role in skeletal muscle defects, the ability of NF‐κB inhibitor dimethylaminoparthenolide (DMAPT) to reverse skeletal muscle defects was examined. Results Neu+ mice showed skeletal muscle defects similar to accelerated aging. Compared with age and sex‐matched wild type mice, Neu+ tumour‐bearing mice had lower grip strength (202 ± 6.9 vs. 179 ± 6.8 g grip force, P = 0.0069) and impaired rotarod performance (108 ± 12.1 vs. 30 ± 3.9 s, P
- Published
- 2021
- Full Text
- View/download PDF
4. TG-interacting factor 1 (Tgif1)-deficiency attenuates bone remodeling and blunts the anabolic response to parathyroid hormone
- Author
-
Hiroaki Saito, Andreas Gasser, Simona Bolamperti, Miki Maeda, Levi Matthies, Katharina Jähn, Courtney L. Long, Hartmut Schlüter, Marcel Kwiatkowski, Vaibhav Saini, Paola Divieti Pajevic, Teresita Bellido, Andre J. van Wijnen, Khalid S. Mohammad, Theresa A. Guise, Hanna Taipaleenmäki, and Eric Hesse
- Subjects
Science - Abstract
Parathyroid hormone (PTH) is used to treat osteoporosis, but its therapeutic mechanism remains unclear. Here, the authors show that Tgif1 is a PTH target gene, and that its deletion impairs the function of osteoblasts and PTH-induced bone formation in mice.
- Published
- 2019
- Full Text
- View/download PDF
5. Translational Strategies to Target Metastatic Bone Disease
- Author
-
Gabriel M. Pagnotti, Trupti Trivedi, and Khalid S. Mohammad
- Subjects
metastatic bone disease ,osteolytic lesions ,osteoblastic lesions ,bone marrow microenvironment ,radiotherapy ,palliative therapy ,Cytology ,QH573-671 - Abstract
Metastatic bone disease is a common and devastating complication to cancer, confounding treatments and recovery efforts and presenting a significant barrier to de-escalating the adverse outcomes associated with disease progression. Despite significant advances in the field, bone metastases remain presently incurable and contribute heavily to cancer-associated morbidity and mortality. Mechanisms associated with metastatic bone disease perpetuation and paralleled disruption of bone remodeling are highlighted to convey how they provide the foundation for therapeutic targets to stem disease escalation. The focus of this review aims to describe the preclinical modeling and diagnostic evaluation of metastatic bone disease as well as discuss the range of therapeutic modalities used clinically and how they may impact skeletal tissue.
- Published
- 2022
- Full Text
- View/download PDF
6. Tip110/SART3 regulates IL-8 expression and predicts the clinical outcomes in melanoma
- Author
-
Khalid Amine Timani, Balázs Győrffy, Ying Liu, Khalid S. Mohammad, and Johnny J. He
- Subjects
Tip110/SART3 ,IL-8 ,Melanoma ,Hypoxia ,TP53, NF-κB ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Tip110, an important regulator of several oncogenic proteins, was significantly downregulated in human metastatic melanoma cells exposed to a hypoxic condition. Therefore, in this study, we set to determine whether differential expression of Tip110 could be an important indicator for melanoma tumorigenesis and metastasis. We found that in melanoma, but not in other cancer types, Tip110 knockdown enhanced significant expression and secretion of IL-8 and melanoma cells invasions. This induction was further potentiated under hypoxia and by inflammatory cytokine and found independent of TNF-α autocrine signaling. We further showed that Tip110 knockdown-mediated IL-8 induction involved IL-8 mRNA stability. Furthermore, the transcriptomic profiling data and survival from 455 melanoma patients demonstrated that the correlation between Tip110 expression and the clinical outcomes in melanoma was stage-dependent. These findings uncover important roles of Tip110 in melanoma tumorigenesis and metastasis through regulation of IL-8 and hope to provide new clues for future therapeutic strategies.
- Published
- 2018
- Full Text
- View/download PDF
7. The Role of TGF-β in Bone Metastases
- Author
-
Trupti Trivedi, Gabriel M. Pagnotti, Theresa A. Guise, and Khalid S. Mohammad
- Subjects
bone metastases ,transforming growth factor-β (TGF-β) ,programmed cell death ligand (PD-L1) ,immune cells ,TGF-β therapeutic targets ,check-point inhibitors ,Microbiology ,QR1-502 - Abstract
Complications associated with advanced cancer are a major clinical challenge and, if associated with bone metastases, worsen the prognosis and compromise the survival of the patients. Breast and prostate cancer cells exhibit a high propensity to metastasize to bone. The bone microenvironment is unique, providing fertile soil for cancer cell propagation, while mineralized bone matrices store potent growth factors and cytokines. Biologically active transforming growth factor β (TGF-β), one of the most abundant growth factors, is released following tumor-induced osteoclastic bone resorption. TGF-β promotes tumor cell secretion of factors that accelerate bone loss and fuel tumor cells to colonize. Thus, TGF-β is critical for driving the feed-forward vicious cycle of tumor growth in bone. Further, TGF-β promotes epithelial-mesenchymal transition (EMT), increasing cell invasiveness, angiogenesis, and metastatic progression. Emerging evidence shows TGF-β suppresses immune responses, enabling opportunistic cancer cells to escape immune checkpoints and promote bone metastases. Blocking TGF-β signaling pathways could disrupt the vicious cycle, revert EMT, and enhance immune response. However, TGF-β’s dual role as both tumor suppressor and enhancer presents a significant challenge in developing therapeutics that target TGF-β signaling. This review presents TGF-β’s role in cancer progression and bone metastases, while highlighting current perspectives on the therapeutic potential of targeting TGF-β pathways.
- Published
- 2021
- Full Text
- View/download PDF
8. Osteocyte-Intrinsic TGF-β Signaling Regulates Bone Quality through Perilacunar/Canalicular Remodeling
- Author
-
Neha S. Dole, Courtney M. Mazur, Claire Acevedo, Justin P. Lopez, David A. Monteiro, Tristan W. Fowler, Bernd Gludovatz, Flynn Walsh, Jenna N. Regan, Sara Messina, Daniel S. Evans, Thomas F. Lang, Bin Zhang, Robert O. Ritchie, Khalid S. Mohammad, and Tamara Alliston
- Subjects
osteocyte ,TGF-β ,bone quality ,perilacunar/canalicular remodeling ,bone fragility ,Biology (General) ,QH301-705.5 - Abstract
Poor bone quality contributes to bone fragility in diabetes, aging, and osteogenesis imperfecta. However, the mechanisms controlling bone quality are not well understood, contributing to the current lack of strategies to diagnose or treat bone quality deficits. Transforming growth factor beta (TGF-β) signaling is a crucial mechanism known to regulate the material quality of bone, but its cellular target in this regulation is unknown. Studies showing that osteocytes directly remodel their perilacunar/canalicular matrix led us to hypothesize that TGF-β controls bone quality through perilacunar/canalicular remodeling (PLR). Using inhibitors and mice with an osteocyte-intrinsic defect in TGF-β signaling (TβRIIocy−/−), we show that TGF-β regulates PLR in a cell-intrinsic manner to control bone quality. Altogether, this study emphasizes that osteocytes are key in executing the biological control of bone quality through PLR, thereby highlighting the fundamental role of osteocyte-mediated PLR in bone homeostasis and fragility.
- Published
- 2017
- Full Text
- View/download PDF
9. Loss of Asxl1 Alters Self-Renewal and Cell Fate of Bone Marrow Stromal Cells, Leading to Bohring-Opitz-like Syndrome in Mice
- Author
-
Peng Zhang, Caihong Xing, Steven D. Rhodes, Yongzheng He, Kai Deng, Zhaomin Li, Fuhong He, Caiying Zhu, Lihn Nguyen, Yuan Zhou, Shi Chen, Khalid S. Mohammad, Theresa A. Guise, Omar Abdel-Wahab, Mingjiang Xu, Qian-Fei Wang, and Feng-Chun Yang
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: De novo ASXL1 mutations are found in patients with Bohring-Opitz syndrome, a disease with severe developmental defects and early childhood mortality. The underlying pathologic mechanisms remain largely unknown. Using Asxl1-targeted murine models, we found that Asxl1 global loss as well as conditional deletion in osteoblasts and their progenitors led to significant bone loss and a markedly decreased number of bone marrow stromal cells (BMSCs) compared with wild-type littermates. Asxl1−/− BMSCs displayed impaired self-renewal and skewed differentiation, away from osteoblasts and favoring adipocytes. RNA-sequencing analysis revealed altered expression of genes involved in cell proliferation, skeletal development, and morphogenesis. Furthermore, gene set enrichment analysis showed decreased expression of stem cell self-renewal gene signature, suggesting a role of Asxl1 in regulating the stemness of BMSCs. Importantly, re-introduction of Asxl1 normalized NANOG and OCT4 expression and restored the self-renewal capacity of Asxl1−/− BMSCs. Our study unveils a pivotal role of ASXL1 in the maintenance of BMSC functions and skeletal development. : In this article, Yang, Wang, and colleagues show that loss of Asxl1 led to multiple skeletal developmental defects, closely reminiscent of Bohring-Opitz syndrome. They identified that the skeletal defects were associated with an impaired self-renewal and skewed lineage commitment of bone marrow stromal cells (BMSCs). These findings indicate a pivotal role of ASXL1 in the maintenance of BMSC functions and skeletal development. Keywords: Bohring-Opitz syndrome, ASXL1 mutation, bone marrow stromal cell, self-renewal and differentiation, skeletal development
- Published
- 2016
- Full Text
- View/download PDF
10. Osteolytic Breast Cancer Causes Skeletal Muscle Weakness in an Immunocompetent Syngeneic Mouse Model
- Author
-
Jenna N. Regan, Carter Mikesell, Steven Reiken, Haifang Xu, Andrew R. Marks, Khalid S. Mohammad, Theresa A. Guise, and David L. Waning
- Subjects
breast cancer ,osteolytic disease ,muscle weakness ,immune competent ,syngeneic tumor model ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Muscle weakness and cachexia are significant paraneoplastic syndromes of many advanced cancers. Osteolytic bone metastases are common in advanced breast cancer and are a major contributor to decreased survival, performance, and quality of life for patients. Pathologic fracture caused by osteolytic cancer in bone (OCIB) leads to a significant (32%) increased risk of death compared to patients without fracture. Since muscle weakness is linked to risk of falls which are a major cause of fracture, we have investigated skeletal muscle response to OCIB. Here, we show that a syngeneic mouse model of OCIB (4T1 mammary tumor cells) leads to cachexia and skeletal muscle weakness associated with oxidation of the ryanodine receptor and calcium (Ca2+) release channel (RyR1). Muscle atrophy follows known pathways via both myostatin signaling and expression of muscle-specific ubiquitin ligases, atrogin-1 and MuRF1. We have identified a mechanism for skeletal muscle weakness due to increased oxidative stress on RyR1 via NAPDH oxidases [NADPH oxidase 2 (Nox2) and NADPH oxidase 4 (Nox4)]. In addition, SMAD3 phosphorylation is higher in muscle from tumor-bearing mice, a critical step in the intracellular signaling pathway that transmits TGFβ signaling to the nucleus. This is the first time that skeletal muscle weakness has been described in a syngeneic model of OCIB and represents a unique model system in which to study cachexia and changes in skeletal muscle.
- Published
- 2017
- Full Text
- View/download PDF
11. Upregulation of SIRT1 Contributes to dmPGE2-dependent Radioprotection of Hematopoietic Stem Cells
- Author
-
Liqiong Liu, Hongge Li, Andrea M. Patterson, P. Artur Plett, Carol H. Sampson, Khalid S. Mohammad, Maegan L. Capitano, Pratibha Singh, Chonghua Yao, Christie M. Orschell, and Louis M. Pelus
- Subjects
General Medicine - Published
- 2022
- Full Text
- View/download PDF
12. Supplementary Figure 1 from Heparin-like Polysaccharides Reduce Osteolytic Bone Destruction and Tumor Growth in a Mouse Model of Breast Cancer Bone Metastasis
- Author
-
Sanna-Maria Käkönen, Olli Kallioniemi, Theresa A. Guise, Merja Perälä, Anne Marjamäki, Marjo Pihlavisto, Liisa Nissinen, Anni Wärri, Jussi M. Halleen, Jukka P. Rissanen, Khalid S. Mohammad, Rami S. Käkönen, and Sirkku Pollari
- Abstract
PDF file - 25K, Supplementary Figure 1. Basal and TGF-b-induced IL-11 production in parental MDA-MB-231 and highly metastatic MDA-MB-231(SA) cells. ***, P < 0.001 (t test).
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figure 2 from Heparin-like Polysaccharides Reduce Osteolytic Bone Destruction and Tumor Growth in a Mouse Model of Breast Cancer Bone Metastasis
- Author
-
Sanna-Maria Käkönen, Olli Kallioniemi, Theresa A. Guise, Merja Perälä, Anne Marjamäki, Marjo Pihlavisto, Liisa Nissinen, Anni Wärri, Jussi M. Halleen, Jukka P. Rissanen, Khalid S. Mohammad, Rami S. Käkönen, and Sirkku Pollari
- Abstract
PDF file - 28K, Supplementary Figure 2. A, the effects of the six most potent siRNA screen hits on IL-11 secretion in MDA-MB-231(SA) cells (n=3). B, the effects of heparan sulfate modifying enzymes on IL-11 secretion in MDA-MB-231(SA) cells (n=4). *, P < 0.05; **, P
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Table 2 from Heparin-like Polysaccharides Reduce Osteolytic Bone Destruction and Tumor Growth in a Mouse Model of Breast Cancer Bone Metastasis
- Author
-
Sanna-Maria Käkönen, Olli Kallioniemi, Theresa A. Guise, Merja Perälä, Anne Marjamäki, Marjo Pihlavisto, Liisa Nissinen, Anni Wärri, Jussi M. Halleen, Jukka P. Rissanen, Khalid S. Mohammad, Rami S. Käkönen, and Sirkku Pollari
- Abstract
XLS file - 49K, Supplementary Table 2. Effects of 196 siRNAs (including 2 siRNAs targeting IL11) on TGF-�-induced IL-11 production in MDA-MB-231(SA) cells. The relative IL-11 concentration values were obtained by dividing the IL-11 concentration by the amount of viable cells in the well (as determined by the CellTiter-Blue assay). The values are shown relative to the average values for the negative control siRNA.
- Published
- 2023
- Full Text
- View/download PDF
15. Data from Heparin-like Polysaccharides Reduce Osteolytic Bone Destruction and Tumor Growth in a Mouse Model of Breast Cancer Bone Metastasis
- Author
-
Sanna-Maria Käkönen, Olli Kallioniemi, Theresa A. Guise, Merja Perälä, Anne Marjamäki, Marjo Pihlavisto, Liisa Nissinen, Anni Wärri, Jussi M. Halleen, Jukka P. Rissanen, Khalid S. Mohammad, Rami S. Käkönen, and Sirkku Pollari
- Abstract
TGF-β regulates several steps in cancer metastasis, including the establishment of bone metastatic lesions. TGF-β is released from bone during osteoclastic bone resorption and it stimulates breast cancer cells to produce osteolytic factors such as interleukin 11 (IL-11). We conducted a cell-based siRNA screen and identified heparan sulfate 6-O-sulfotransferase 2 (HS6ST2) as a critical gene for TGF-β–induced IL-11 production in highly bone metastatic MDA-MB-231(SA) breast cancer cells. HS6ST2 attaches sulfate groups to glucosamine residues in heparan sulfate glycosaminoglycans. We subsequently showed how heparin and a high-molecular-weight Escherichia coli K5-derived heparin-like polysaccharide (K5-NSOS) inhibited TGF-β–induced IL-11 production in MDA-MB-231(SA) cells. In addition, K5-NSOS inhibited bone resorption activity of human osteoclasts in vitro. We evaluated the therapeutic potential of K5-NSOS and fragmin in a mouse model of breast cancer bone metastasis. MDA-MB-231(SA) cells were inoculated into the left cardiac ventricle of athymic nude mice which were treated with fragmin, K5-NSOS, or vehicle once a day for four weeks. Both heparin-like glycosaminoglycans inhibited weight reduction, decreased osteolytic lesion area, and reduced tumor burden in bone. In conclusion, our data imply novel mechanisms involved in TGF-β induction and support the critical role of heparan sulfate glycosaminoglycans in cancer metastasis as well as indicate that K5-NSOS is a potential antimetastatic and antiresorptive agent for cancer therapy. This study illustrates the potential to translate in vitro siRNA screening results toward in vivo therapeutic concepts. Mol Cancer Res; 10(5); 597–604. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Table 1 from Heparin-like Polysaccharides Reduce Osteolytic Bone Destruction and Tumor Growth in a Mouse Model of Breast Cancer Bone Metastasis
- Author
-
Sanna-Maria Käkönen, Olli Kallioniemi, Theresa A. Guise, Merja Perälä, Anne Marjamäki, Marjo Pihlavisto, Liisa Nissinen, Anni Wärri, Jussi M. Halleen, Jukka P. Rissanen, Khalid S. Mohammad, Rami S. Käkönen, and Sirkku Pollari
- Abstract
PDF file - 6K, Supplementary Table 1. Primers and probes used for quantitative RT-PCR.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary Figure 3 from Heparin-like Polysaccharides Reduce Osteolytic Bone Destruction and Tumor Growth in a Mouse Model of Breast Cancer Bone Metastasis
- Author
-
Sanna-Maria Käkönen, Olli Kallioniemi, Theresa A. Guise, Merja Perälä, Anne Marjamäki, Marjo Pihlavisto, Liisa Nissinen, Anni Wärri, Jussi M. Halleen, Jukka P. Rissanen, Khalid S. Mohammad, Rami S. Käkönen, and Sirkku Pollari
- Abstract
PDF file - 38K, Supplementary Figure 3. A, expression of HS6ST2 mRNA in MDA-MB-231(SA) versus parental MDA-MB-231 cells. B, validation of the knockdown efficacies of siRNAs by quantitative RT-PCR.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary figure 1 from Bidirectional Notch Signaling and Osteocyte-Derived Factors in the Bone Marrow Microenvironment Promote Tumor Cell Proliferation and Bone Destruction in Multiple Myeloma
- Author
-
Teresita Bellido, G. David Roodman, Lilian I. Plotkin, Khalid S. Mohammad, Toshiyuki Yoneda, Nadia Carlesso, John M. Chirgwin, Masahiro Hiasa, Meloney D. Cregor, Judith Anderson, and Jesus Delgado-Calle
- Abstract
Supplementary figure 1. MM cells induce apoptosis and activate Notch signaling in osteocytes.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplemental Figure S1 from Bone-Induced Expression of Integrin β3 Enables Targeted Nanotherapy of Breast Cancer Metastases
- Author
-
Katherine N. Weilbaecher, Gregory M. Lanza, Theresa A. Guise, Khalid S. Mohammad, David L. Waning, James A. J. Fitzpatrick, Joshua Novack, Elizabeth Cordell, Gabriel H. Lukaszewicz, Graham A. Colditz, Thomas Walsh, Deborah V. Novack, Yalin Xu, Xinming Su, Dipanjan Pan, Grace Hu, Xiaoxia Yang, Anne H. Schmieder, Gregory C. Fox, Alison K. Esser, and Michael H. Ross
- Abstract
Immunohistochemistry and flow cytometry of tumor cells for integrin expression
- Published
- 2023
- Full Text
- View/download PDF
20. Supplemental figure legend from Cell Adhesion Molecule CD166 Drives Malignant Progression and Osteolytic Disease in Multiple Myeloma
- Author
-
Edward F. Srour, Attaya Suvannasankha, John Chirgwin, Melissa A. Kacena, Rafat Abonour, Helmut Hanenberg, Christophe Machal, Angelo A. Cardoso, Yinghua Cheng, Bradley Poteat, Colin Crean, Hao Wu, Khalid S. Mohammad, and Linlin Xu
- Abstract
Supplemental figure legend
- Published
- 2023
- Full Text
- View/download PDF
21. Supplemental Information from Bone-Induced Expression of Integrin β3 Enables Targeted Nanotherapy of Breast Cancer Metastases
- Author
-
Katherine N. Weilbaecher, Gregory M. Lanza, Theresa A. Guise, Khalid S. Mohammad, David L. Waning, James A. J. Fitzpatrick, Joshua Novack, Elizabeth Cordell, Gabriel H. Lukaszewicz, Graham A. Colditz, Thomas Walsh, Deborah V. Novack, Yalin Xu, Xinming Su, Dipanjan Pan, Grace Hu, Xiaoxia Yang, Anne H. Schmieder, Gregory C. Fox, Alison K. Esser, and Michael H. Ross
- Abstract
Supplemental Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary figure 3 from Bidirectional Notch Signaling and Osteocyte-Derived Factors in the Bone Marrow Microenvironment Promote Tumor Cell Proliferation and Bone Destruction in Multiple Myeloma
- Author
-
Teresita Bellido, G. David Roodman, Lilian I. Plotkin, Khalid S. Mohammad, Toshiyuki Yoneda, Nadia Carlesso, John M. Chirgwin, Masahiro Hiasa, Meloney D. Cregor, Judith Anderson, and Jesus Delgado-Calle
- Abstract
Supplementary figure 3. Osteocytes regulate cell proliferation and Notch receptor expression in MM cells.
- Published
- 2023
- Full Text
- View/download PDF
23. Data from Bidirectional Notch Signaling and Osteocyte-Derived Factors in the Bone Marrow Microenvironment Promote Tumor Cell Proliferation and Bone Destruction in Multiple Myeloma
- Author
-
Teresita Bellido, G. David Roodman, Lilian I. Plotkin, Khalid S. Mohammad, Toshiyuki Yoneda, Nadia Carlesso, John M. Chirgwin, Masahiro Hiasa, Meloney D. Cregor, Judith Anderson, and Jesus Delgado-Calle
- Abstract
In multiple myeloma, an overabundance of monoclonal plasma cells in the bone marrow induces localized osteolytic lesions that rarely heal due to increased bone resorption and suppressed bone formation. Matrix-embedded osteocytes comprise more than 95% of bone cells and are major regulators of osteoclast and osteoblast activity, but their contribution to multiple myeloma growth and bone disease is unknown. Here, we report that osteocytes in a mouse model of human MM physically interact with multiple myeloma cells in vivo, undergo caspase-3–dependent apoptosis, and express higher RANKL (TNFSF11) and sclerostin levels than osteocytes in control mice. Mechanistic studies revealed that osteocyte apoptosis was initiated by multiple myeloma cell-mediated activation of Notch signaling and was further amplified by multiple myeloma cell-secreted TNF. The induction of apoptosis increased osteocytic Rankl expression, the osteocytic Rankl/Opg (TNFRSF11B) ratio, and the ability of osteocytes to attract osteoclast precursors to induce local bone resorption. Furthermore, osteocytes in contact with multiple myeloma cells expressed high levels of Sost/sclerostin, leading to a reduction in Wnt signaling and subsequent inhibition of osteoblast differentiation. Importantly, direct contact between osteocytes and multiple myeloma cells reciprocally activated Notch signaling and increased Notch receptor expression, particularly Notch3 and 4, stimulating multiple myeloma cell growth. These studies reveal a previously unknown role for bidirectional Notch signaling that enhances MM growth and bone disease, suggesting that targeting osteocyte-multiple myeloma cell interactions through specific Notch receptor blockade may represent a promising treatment strategy in multiple myeloma. Cancer Res; 76(5); 1089–100. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
24. Supplemental figures 1-7 from Cell Adhesion Molecule CD166 Drives Malignant Progression and Osteolytic Disease in Multiple Myeloma
- Author
-
Edward F. Srour, Attaya Suvannasankha, John Chirgwin, Melissa A. Kacena, Rafat Abonour, Helmut Hanenberg, Christophe Machal, Angelo A. Cardoso, Yinghua Cheng, Bradley Poteat, Colin Crean, Hao Wu, Khalid S. Mohammad, and Linlin Xu
- Abstract
S1: Distribution of CD166 mRNA expression across 30 multiple myeloma and 22 breast cancer cell lines were analyzed; S2: The percentage of CD166+ cells within H929 cells from BM-homed cells were examined flow cytometrically; S3: Data are representative of 2 separate experiments (mean{plus minus}SEM, N = 6mice/group/experiment, each assayed individually); S5: Data represent 3 separate experiments done in triplicates for each group and are expressed as mean{plus minus} SEM, *P
- Published
- 2023
- Full Text
- View/download PDF
25. Data from Bone-Induced Expression of Integrin β3 Enables Targeted Nanotherapy of Breast Cancer Metastases
- Author
-
Katherine N. Weilbaecher, Gregory M. Lanza, Theresa A. Guise, Khalid S. Mohammad, David L. Waning, James A. J. Fitzpatrick, Joshua Novack, Elizabeth Cordell, Gabriel H. Lukaszewicz, Graham A. Colditz, Thomas Walsh, Deborah V. Novack, Yalin Xu, Xinming Su, Dipanjan Pan, Grace Hu, Xiaoxia Yang, Anne H. Schmieder, Gregory C. Fox, Alison K. Esser, and Michael H. Ross
- Abstract
Bone metastases occur in approximately 70% of metastatic breast cancer patients, often leading to skeletal injuries. Current treatments are mainly palliative and underscore the unmet clinical need for improved therapies. In this study, we provide preclinical evidence for an antimetastatic therapy based on targeting integrin β3 (β3), which is selectively induced on breast cancer cells in bone by the local bone microenvironment. In a preclinical model of breast cancer, β3 was strongly expressed on bone metastatic cancer cells, but not primary mammary tumors or visceral metastases. In tumor tissue from breast cancer patients, β3 was significantly elevated on bone metastases relative to primary tumors from the same patient (n = 42). Mechanistic investigations revealed that TGFβ signaling through SMAD2/SMAD3 was necessary for breast cancer induction of β3 within the bone. Using a micelle-based nanoparticle therapy that recognizes integrin αvβ3 (αvβ3-MPs of ∼12.5 nm), we demonstrated specific localization to breast cancer bone metastases in mice. Using this system for targeted delivery of the chemotherapeutic docetaxel, we showed that bone tumor burden could be reduced significantly with less bone destruction and less hepatotoxicity compared with equimolar doses of free docetaxel. Furthermore, mice treated with αvβ3-MP-docetaxel exhibited a significant decrease in bone-residing tumor cell proliferation compared with free docetaxel. Taken together, our results offer preclinical proof of concept for a method to enhance delivery of chemotherapeutics to breast cancer cells within the bone by exploiting their selective expression of integrin αvβ3 at that metastatic site. Cancer Res; 77(22); 6299–312. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary figure 2 from Bidirectional Notch Signaling and Osteocyte-Derived Factors in the Bone Marrow Microenvironment Promote Tumor Cell Proliferation and Bone Destruction in Multiple Myeloma
- Author
-
Teresita Bellido, G. David Roodman, Lilian I. Plotkin, Khalid S. Mohammad, Toshiyuki Yoneda, Nadia Carlesso, John M. Chirgwin, Masahiro Hiasa, Meloney D. Cregor, Judith Anderson, and Jesus Delgado-Calle
- Abstract
Supplementary figure 2. MM cells increase Rankl mRNA levels in osteocytes.
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary figure legends from Bidirectional Notch Signaling and Osteocyte-Derived Factors in the Bone Marrow Microenvironment Promote Tumor Cell Proliferation and Bone Destruction in Multiple Myeloma
- Author
-
Teresita Bellido, G. David Roodman, Lilian I. Plotkin, Khalid S. Mohammad, Toshiyuki Yoneda, Nadia Carlesso, John M. Chirgwin, Masahiro Hiasa, Meloney D. Cregor, Judith Anderson, and Jesus Delgado-Calle
- Abstract
Supplementary figure legends
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary methods from Bidirectional Notch Signaling and Osteocyte-Derived Factors in the Bone Marrow Microenvironment Promote Tumor Cell Proliferation and Bone Destruction in Multiple Myeloma
- Author
-
Teresita Bellido, G. David Roodman, Lilian I. Plotkin, Khalid S. Mohammad, Toshiyuki Yoneda, Nadia Carlesso, John M. Chirgwin, Masahiro Hiasa, Meloney D. Cregor, Judith Anderson, and Jesus Delgado-Calle
- Abstract
Supplementary methods
- Published
- 2023
- Full Text
- View/download PDF
29. Supplemental Table 1 from Cell Adhesion Molecule CD166 Drives Malignant Progression and Osteolytic Disease in Multiple Myeloma
- Author
-
Edward F. Srour, Attaya Suvannasankha, John Chirgwin, Melissa A. Kacena, Rafat Abonour, Helmut Hanenberg, Christophe Machal, Angelo A. Cardoso, Yinghua Cheng, Bradley Poteat, Colin Crean, Hao Wu, Khalid S. Mohammad, and Linlin Xu
- Abstract
Supplemental table 1
- Published
- 2023
- Full Text
- View/download PDF
30. Data from Cell Adhesion Molecule CD166 Drives Malignant Progression and Osteolytic Disease in Multiple Myeloma
- Author
-
Edward F. Srour, Attaya Suvannasankha, John Chirgwin, Melissa A. Kacena, Rafat Abonour, Helmut Hanenberg, Christophe Machal, Angelo A. Cardoso, Yinghua Cheng, Bradley Poteat, Colin Crean, Hao Wu, Khalid S. Mohammad, and Linlin Xu
- Abstract
Multiple myeloma is incurable once osteolytic lesions have seeded at skeletal sites, but factors mediating this deadly pathogenic advance remain poorly understood. Here, we report evidence of a major role for the cell adhesion molecule CD166, which we discovered to be highly expressed in multiple myeloma cell lines and primary bone marrow cells from patients. CD166+ multiple myeloma cells homed more efficiently than CD166− cells to the bone marrow of engrafted immunodeficient NSG mice. CD166 silencing in multiple myeloma cells enabled longer survival, a smaller tumor burden, and less osteolytic lesions, as compared with mice bearing control cells. CD166 deficiency in multiple myeloma cell lines or CD138+ bone marrow cells from multiple myeloma patients compromised their ability to induce bone resorption in an ex vivo organ culture system. Furthermore, CD166 deficiency in multiple myeloma cells also reduced the formation of osteolytic disease in vivo after intratibial engraftment. Mechanistic investigation revealed that CD166 expression in multiple myeloma cells inhibited osteoblastogenesis of bone marrow–derived osteoblast progenitors by suppressing Runx2 gene expression. Conversely, CD166 expression in multiple myeloma cells promoted osteoclastogenesis by activating TRAF6-dependent signaling pathways in osteoclast progenitors. Overall, our results define CD166 as a pivotal director in multiple myeloma cell homing to the bone marrow and multiple myeloma progression, rationalizing its further study as a candidate therapeutic target for multiple myeloma treatment. Cancer Res; 76(23); 6901–10. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
31. Data from Stable Overexpression of Smad7 in Human Melanoma Cells Impairs Bone Metastasis
- Author
-
Alain Mauviel, Theresa A. Guise, Lionel Larue, Véronique Delmas, Jocelyne André, Maryla Niewolna, Flavie Luciani, Pierrick Fournier, Christopher R. McKenna, Khalid S. Mohammad, and Delphine Javelaud
- Abstract
Melanoma has a propensity to metastasize to bone, where it is exposed to high concentrations of transforming growth factor-β (TGF-β). Because TGF-β promotes bone metastases from other solid tumors, such as breast cancer, we tested the role of TGF-β in melanoma metastases to bone. 1205Lu melanoma cells, stably transfected to overexpress the natural TGF-β/Smad signaling inhibitor Smad7, were studied in an experimental model of bone metastasis whereby tumor cells are inoculated into the left cardiac ventricle of nude mice. All mice bearing parental and mock-transfected 1205Lu cells developed osteolytic bone metastases 5 weeks post-tumor inoculation. Mice bearing 1205Lu-Smad7 tumors had significantly less osteolysis on radiographs and longer survival compared with parental and mock-transfected 1205Lu mice. To determine if the reduced bone metastases observed in mice bearing 1205Lu-Smad7 clones was due to reduced expression of TGF-β target genes known to enhance metastases to bone from breast cancer cells, we analyzed gene expression of osteolytic factors, parathyroid hormone-related protein (PTHrP) and interleukin-11 (IL-11), the chemotactic receptor CXCR4, and osteopontin in 1205Lu cells. Quantitative reverse transcription-PCR analysis indicated that PTHrP, IL-11, CXCR4, and osteopontin mRNA steady-state levels were robustly increased in response to TGF-β and that Smad7 and the TβRI small-molecule inhibitor, SB431542, prevented such induction. In addition, 1205Lu-Smad7 bone metastases expressed significantly lower levels of IL-11, connective tissue growth factor, and PTHrP. These data suggest that TGF-β promotes osteolytic bone metastases due to melanoma by stimulating the expression of prometastatic factors via the Smad pathway. Blockade of TGF-β signaling may be an effective treatment for melanoma metastasis to bone. [Cancer Res 2007;67(5):2317–24]
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Figure 1 from Stable Overexpression of Smad7 in Human Melanoma Cells Impairs Bone Metastasis
- Author
-
Alain Mauviel, Theresa A. Guise, Lionel Larue, Véronique Delmas, Jocelyne André, Maryla Niewolna, Flavie Luciani, Pierrick Fournier, Christopher R. McKenna, Khalid S. Mohammad, and Delphine Javelaud
- Abstract
Supplementary Figure 1 from Stable Overexpression of Smad7 in Human Melanoma Cells Impairs Bone Metastasis
- Published
- 2023
- Full Text
- View/download PDF
33. Supplementary Figure 1 Legend from Stable Overexpression of Smad7 in Human Melanoma Cells Impairs Bone Metastasis
- Author
-
Alain Mauviel, Theresa A. Guise, Lionel Larue, Véronique Delmas, Jocelyne André, Maryla Niewolna, Flavie Luciani, Pierrick Fournier, Christopher R. McKenna, Khalid S. Mohammad, and Delphine Javelaud
- Abstract
Supplementary Figure 1 Legend from Stable Overexpression of Smad7 in Human Melanoma Cells Impairs Bone Metastasis
- Published
- 2023
- Full Text
- View/download PDF
34. Low-Magnitude Mechanical Signals Combined with Zoledronic Acid Reduce Musculoskeletal Weakness and Adiposity in Estrogen-Deprived Mice
- Author
-
Gabriel M. Pagnotti, Trupti Trivedi, Laura E. Wright, Sutha K. John, Sreemala Murthy, Ryan R. Pattyn, Monte S. Willis, Yun She, Sukanya Suresh, William R. Thompson, Clinton T. Rubin, Khalid S. Mohammad, and Theresa A. Guise
- Subjects
Article - Abstract
Combination treatment of Low-Intensity Vibration (LIV) with zoledronic acid (ZA) was hypothesized to preserve bone mass and muscle strength while reducing adipose tissue accrual associated with complete estrogen (E2)-deprivation in young and skeletally mature mice. Complete E2-deprivation (surgical-ovariectomy (OVX) and daily injection of aromatase inhibitor (AI) letrozole) were performed on 8-week-old C57BL/6 female mice for 4 weeks following commencement of LIV administration or control (no LIV), for 28 weeks. Additionally, 16-week-old C57BL/6 female E2-deprived mice were administered ±LIV twice daily and supplemented with ±ZA (2.5 ng/kg/week). By week 28, lean tissue mass quantified by dual-energy X-ray absorptiometry was increased in younger OVX/AI+LIV(y) mice, with increased myofiber cross-sectional area of quadratus femorii. Grip strength was greater in OVX/AI+LIV(y) mice than OVX/AI(y) mice. Fat mass remained lower in OVX/AI+LIV(y) mice throughout the experiment compared with OVX/AI(y) mice. OVX/AI+LIV(y) mice exhibited increased glucose tolerance and reduced leptin and free fatty acids than OVX/AI(y) mice. Trabecular bone volume fraction and connectivity density increased in the vertebrae of OVX/AI+LIV(y) mice compared to OVX/AI(y) mice; however, this effect was attenuated in the older cohort of E2-deprived mice, specifically in OVX/AI+ZA mice, requiring combined LIV with ZA to increase trabecular bone volume and strength. Similar improvements in cortical bone thickness and cross-sectional area of the femoral mid-diaphysis were observed in OVX/AI+LIV+ZA mice, resulting in greater fracture resistance. Our findings demonstrate that the combination of mechanical signals in the form of LIV and anti-resorptive therapy via ZA improve vertebral trabecular bone and femoral cortical bone, increase lean mass, and reduce adiposity in mice undergoing complete E2-deprivation.One Sentence Summary:Low-magnitude mechanical signals with zoledronic acid suppressed bone and muscle loss and adiposity in mice undergoing complete estrogen deprivation.Translational RelevancePostmenopausal patients with estrogen receptor-positive breast cancer treated with aromatase inhibitors to reduce tumor progression experience deleterious effects to bone and muscle subsequently develop muscle weakness, bone fragility, and adipose tissue accrual. Bisphosphonates (i.e., zoledronic acid) prescribed to inhibit osteoclast-mediated bone resorption are effective in preventing bone loss but may not address the non-skeletal effects of muscle weakness and fat accumulation that contribute to patient morbidity. Mechanical signals, typically delivered to the musculoskeletal system during exercise/physical activity, are integral for maintaining bone and muscle health; however, patients undergoing treatments for breast cancer often experience decreased physical activity which further accelerates musculoskeletal degeneration. Low-magnitude mechanical signals, in the form of low-intensity vibrations, generate dynamic loading forces similar to those derived from skeletal muscle contractility. As an adjuvant to existing treatment strategies, low-intensity vibrations may preserve or rescue diminished bone and muscle degraded by breast cancer treatment.
- Published
- 2023
- Full Text
- View/download PDF
35. Comparison of electric motors used in electric vehicle propulsion system
- Author
-
Khalid S. Mohammad and Aqeel S. Jaber
- Subjects
Control and Optimization ,Electric vehicles ,Computer Networks and Communications ,Hardware and Architecture ,Signal Processing ,Electrical and Electronic Engineering ,Induction motor ,Switched reluctance motor artificial intelligent ,Information Systems ,DC motors ,Permanent magnet brushless motors - Abstract
Now days, it is vital to use electric vehicles (EVs) instead of traditional cars with internal combustion engines (ICEs) in order to reduce the high level of pollution in the environment, and many researchers are investigating the possible improvements on these vehicles. The main component of EVs is the electric motor and the selection of a motor with high efficiency, excellent dynamic response and high starting torque has a strong effect on the performance of EVs. In addition to that a reasonable price for the electric motor is required. This work focuses on the selection of the most suitable electric motor for EVs. Therefore a detailed study to compare between the performance of the major types of electric motors that are used in EVs is addressed in this paper. The results of this comparative study is tabulated and by careful consideration for all these results, the appropriate electric motor for EVs has been chosen. From the other hand, the artificial intelligent (AI) techniques play a crucial role in the EVs technologies, and several kinds of AI techniques used in EVs applications are overviewed in this work.
- Published
- 2022
36. Increased S1P expression in osteoclasts enhances bone formation in an animal model of Paget's disease
- Author
-
Kazuaki Miyagawa, Theresa A. Guise, Khalid S. Mohammad, Noriyoshi Kurihara, Daniela N. Petrusca, Jolene J. Windle, Yuki Nagata, Yasuhisa Ohata, G. David Roodman, and Gabriel M. Pagnotti
- Subjects
0301 basic medicine ,Agonist ,Genetically modified mouse ,Male ,medicine.drug_class ,medicine.medical_treatment ,Immunoblotting ,Osteoclasts ,Enzyme-Linked Immunosorbent Assay ,Biochemistry ,S1P ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,SphK‐1 ,Osteogenesis ,Sphingosine ,medicine ,Animals ,Humans ,Bone formation ,Phosphorylation ,Molecular Biology ,S1PR3 ,Sphingosine-1-Phosphate Receptors ,Research Articles ,bone formation ,Paget's bone disease ,Interleukin-6 ,Growth factor ,Antagonist ,Cell Biology ,Osteitis Deformans ,Molecular biology ,Immunohistochemistry ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Female ,Lysophospholipids ,Research Article - Abstract
Paget's disease (PD) is characterized by increased numbers of abnormal osteoclasts (OCLs) that drive exuberant bone formation, but the mechanisms responsible for the increased bone formation remain unclear. We previously reported that OCLs from 70% of PD patients express measles virus nucleocapsid protein (MVNP), and that transgenic mice with targeted expression of MVNP in OCLs (MVNP mice) develop bone lesions and abnormal OCLs characteristic of PD. In this report, we examined if OCL‐derived sphingosine‐1‐phosphate (S1P) contributed to the abnormal bone formation in PD, since OCL‐derived S1P can act as a coupling factor to increase normal bone formation via binding S1P‐receptor‐3 (S1PR3) on osteoblasts (OBs). We report that OCLs from MVNP mice and PD patients expressed high levels of sphingosine kinase‐1 (SphK‐1) compared with wild‐type (WT) mouse and normal donor OCLs. SphK‐1 production by MVNP‐OCLs was interleukin‐6 (IL‐6)‐dependent since OCLs from MVNP/IL‐6 −/− mice expressed lower levels of SphK‐1. Immunohistochemistry of bone biopsies from a normal donor, a PD patient, WT and MVNP mice confirmed increased expression levels of SphK‐1 in OCLs and S1PR3 in OBs of the PD patient and MVNP mice compared with normal donor and WT mice. Further, MVNP‐OCLs cocultured with OBs from MVNP or WT mice increased OB‐S1PR3 expression and enhanced expression of OB differentiation markers in MVNP‐OBs precursors compared with WT‐OBs, which was mediated by IL‐6 and insulin‐like growth factor 1 secreted by MVNP‐OCLs. Finally, the addition of an S1PR3 antagonist (VPC23019) to WT or MVNP‐OBs treated with WT and MVNP‐OCL‐conditioned media (CM) blocked enhanced OB differentiation of MVNP‐OBs treated with MVNP‐OCL‐CM. In contrast, the addition of the SIPR3 agonist, VPC24191, to the cultures enhanced osterix and Col‐1A expression in MVNP‐OBs treated with MVNP‐OCL‐CM compared with WT‐OBs treated with WT‐OCL‐CM. These results suggest that IL‐6 produced by PD‐OCLs increases S1P in OCLs and S1PR3 on OBs, to increase bone formation in PD., Model for the effects of sphingosine kinase‐1/sphingosine‐1‐phosphate/S1P‐receptor‐3 (SphK‐1/S1P/S1PR3) in abnormal bone remodeling in Paget's disease (PD). Measles virus nucleocapsid protein (MVNP) in PD‐osteoclasts (OCLs) induces interleukin‐6 (IL‐6), which upregulates insulin‐like growth factor 1 (IGF‐1) and Sphk1. SphK‐1 enhances S1P levels in OCLs, and IL‐6 and IGF‐1 increase S1PR3 on osteoblast (OBs). S1P then increases OB differentiation and bone formation via enhanced S1PR3 expression on OBs.
- Published
- 2020
37. A Single Radioprotective Dose of Prostaglandin E2 Blocks Irradiation-Induced Apoptotic Signaling and Early Cycling of Hematopoietic Stem Cells
- Author
-
Jonathan Hoggatt, Khalid S. Mohammad, Andrea M. Patterson, Carol H. Sampson, Liqiong Liu, Maegan L. Capitano, Hongge Li, P. Artur Plett, Pratibha Singh, Christie M. Orschell, and Louis M. Pelus
- Subjects
0301 basic medicine ,Hematopoietic stem cell ,Cell Biology ,Biology ,Cell cycle ,Biochemistry ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Bone marrow suppression ,Genetics ,medicine ,Cancer research ,Bone marrow ,Stem cell ,Prostaglandin E2 ,Progenitor cell ,030217 neurology & neurosurgery ,Developmental Biology ,medicine.drug - Abstract
Summary Ionizing radiation exposure results in acute and delayed bone marrow suppression. Treatment of mice with 16,16-dimethyl prostaglandin E2 (dmPGE2) prior to lethal ionizing radiation (IR) facilitates survival, but the cellular and molecular mechanisms are unclear. In this study we show that dmPGE2 attenuates loss and enhances recovery of bone marrow cellularity, corresponding to a less severe hematopoietic stem cell nadir, and significantly preserves long-term repopulation capacity and progenitor cell function. Mechanistically, dmPGE2 suppressed hematopoietic stem cell (HSC) proliferation through 24 h post IR, which correlated with fewer DNA double-strand breaks and attenuation of apoptosis, mitochondrial compromise, oxidative stress, and senescence. RNA sequencing of HSCs at 1 h and 24 h post IR identified a predominant interference with IR-induced p53-downstream gene expression at 1 h, and confirmed the suppression of IR-induced cell-cycle genes at 24 h. These data identify mechanisms of dmPGE2 radioprotection and its potential role as a medical countermeasure against radiation exposure.
- Published
- 2020
- Full Text
- View/download PDF
38. CXCR4 expression in the bone marrow microenvironment is required for hematopoietic stem and progenitor cell maintenance and early hematopoietic regeneration after myeloablation
- Author
-
Khalid S. Mohammad, Pratibha Singh, and Louis M. Pelus
- Subjects
0301 basic medicine ,Receptors, CXCR4 ,Transplantation Conditioning ,Stromal cell ,Bone Marrow Cells ,Stem cell factor ,Biology ,CXCR4 ,Article ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Bone Marrow ,medicine ,Animals ,Progenitor cell ,Mesenchymal stem cell ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematopoietic Stem Cells ,Chemokine CXCL12 ,Cell biology ,Transplantation ,030104 developmental biology ,medicine.anatomical_structure ,Molecular Medicine ,Bone marrow ,030217 neurology & neurosurgery ,Developmental Biology ,Homing (hematopoietic) - Abstract
The bone marrow (BM) microenvironment/niche plays a key role in regulating hematopoietic stem and progenitor cell (HSPC) activities; however, mechanisms regulating niche cell function are not well understood. In this study, we show that niche intrinsic expression of the CXCR4 chemokine receptor critically regulates HSPC maintenance during steady state, and promotes early hematopoietic regeneration after myeloablative irradiation. At steady state, chimeric mice with wild-type (WT) HSPC and marrow stroma that lack CXCR4 show decreased HSPC quiescence, and their repopulation capacity was markedly reduced. Mesenchymal stromal cells (MSC) were significantly reduced in the BM of CXCR4 deficient mice, which was accompanied by decreased levels of the HSPC supporting factors stromal cell-derived factor-1 (SDF-1) and stem cell factor (SCF). CXCR4 also plays a crucial role in survival and restoration of BM stromal cells after myeloablative irradiation, where the loss of BM stromal cells was more severe in CXCR4-deficient mice compared to WT mice. In addition, transplantation of WT donor HSPC into CXCR4-deficient recipient mice demonstrated reduced HSPC homing and early hematopoietic reconstitution. We found that CXCR4 signaling attenuates irradiation-induced BM stromal cell loss by upregulating the expression of the antiapoptotic protein Survivin via the PI3K pathway. Our study suggests that SDF-1-CXCR4 signaling in the stromal microenvironment cells plays a crucial role in maintenance of HSPCs during homeostasis, and promotes niche regeneration and early hematopoietic reconstitution after transplantation. Modulation of CXCR4 signaling in the HSPC microenvironment could be a means to enhance hematopoietic recovery after clinical hematopoietic cell transplantation.
- Published
- 2020
- Full Text
- View/download PDF
39. TG-interacting factor 1 (Tgif1)-deficiency attenuates bone remodeling and blunts the anabolic response to parathyroid hormone
- Author
-
Vaibhav Saini, Khalid S. Mohammad, Levi Matthies, Courtney L. Long, Hiroaki Saito, Andre J. van Wijnen, Simona Bolamperti, Hartmut Schlüter, Miki Maeda, Marcel Kwiatkowski, Paola Divieti Pajevic, Andreas Gasser, Katharina Jähn, Hanna Taipaleenmaki, Theresa A. Guise, Teresita Bellido, and Eric Hesse
- Subjects
0301 basic medicine ,Anabolism ,Osteoporosis ,Osteoclasts ,General Physics and Astronomy ,Parathyroid hormone ,Semaphorins ,02 engineering and technology ,Bone remodeling ,Anabolic Agents ,lcsh:Science ,Wnt Signaling Pathway ,Multidisciplinary ,Chemistry ,Cell Differentiation ,Osteoblast ,Organ Size ,021001 nanoscience & nanotechnology ,3. Good health ,medicine.anatomical_structure ,Parathyroid Hormone ,Intercellular Signaling Peptides and Proteins ,Bone Remodeling ,0210 nano-technology ,hormones, hormone substitutes, and hormone antagonists ,medicine.medical_specialty ,endocrine system ,Science ,Bone and Bones ,Article ,General Biochemistry, Genetics and Molecular Biology ,Bone resorption ,03 medical and health sciences ,Semaphorin ,Internal medicine ,medicine ,Animals ,Secretion ,Adaptor Proteins, Signal Transducing ,Glycoproteins ,Osteoblasts ,General Chemistry ,medicine.disease ,Mice, Inbred C57BL ,Repressor Proteins ,Transcription Factor AP-1 ,030104 developmental biology ,Endocrinology ,lcsh:Q ,Gene Deletion - Abstract
Osteoporosis is caused by increased bone resorption and decreased bone formation. Intermittent administration of a fragment of Parathyroid hormone (PTH) activates osteoblast-mediated bone formation and is used in patients with severe osteoporosis. However, the mechanisms by which PTH elicits its anabolic effect are not fully elucidated. Here we show that the absence of the homeodomain protein TG-interacting factor 1 (Tgif1) impairs osteoblast differentiation and activity, leading to a reduced bone formation. Deletion of Tgif1 in osteoblasts and osteocytes decreases bone resorption due to an increased secretion of Semaphorin 3E (Sema3E), an osteoclast-inhibiting factor. Tgif1 is a PTH target gene and PTH treatment failed to increase bone formation and bone mass in Tgif1-deficient mice. Thus, our study identifies Tgif1 as a novel regulator of bone remodeling and an essential component of the PTH anabolic action. These insights contribute to a better understanding of bone metabolism and the anabolic function of PTH., Parathyroid hormone (PTH) is used to treat osteoporosis, but its therapeutic mechanism remains unclear. Here, the authors show that Tgif1 is a PTH target gene, and that its deletion impairs the function of osteoblasts and PTH-induced bone formation in mice.
- Published
- 2019
- Full Text
- View/download PDF
40. Ossabaw Pig Demonstrates Detrusor Fibrosis and Detrusor Underactivity Associated with Oxidative Stress in Metabolic Syndrome
- Author
-
Khalid S. Mohammad, Mouhamad Alloosh, Charles R. Powell, James Byrd, Michael Sturek, Joshua D. Roth, Ragini Vittal, and Albert Kim
- Subjects
medicine.medical_specialty ,Swine ,Urinary system ,Pig bladder ,Urine ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Fibrosis ,Internal medicine ,Urinary Bladder, Underactive ,medicine ,TBARS ,Animals ,Original Research ,Metabolic Syndrome ,General Veterinary ,business.industry ,medicine.disease ,Rats ,Oxidative Stress ,Endocrinology ,Blood pressure ,Metabolic syndrome ,business ,Oxidative stress - Abstract
Metabolic Syndrome (MetS) has detrimental effects on the bladder, including detrusor underactivity. The progression and mechanism of disease are poorly understood. A swine model for diabetic bladder dysfunction (DBD) was established because of the pig's human-sized bladder and its ability to develop MetS by dietary modification alone. The hypothesis of this study is that this swine model will demonstrate oxidative stress associated with MetS, which contributes to both bladder fibrosis and detrusor underactivity (DU). Ossabaw pigs underwent dietary modification consisting of a hypercaloric, atherogenic diet for 10 mo to induce MetS, and were compared with a group of control (lean) pigs. Urodynamic studies were performed in both groups to confirm DU. Thiobarbituric acid reactive substances (TBARS) detected in the urine were used to measure oxidative stress activity in the urinary tract, and urinary IL17a was used to detect profibrotic activity. MetS was confirmed by assessing body weight, blood pressure, glucose tolerance, total cholesterol, and triglycerides. The MetS group exhibited an increase in the relative levels of urinary TBARS and IL17a. Bladder pressures at capacity were lower in the MetS group, suggesting DU. Histologic analysis of a cohort of control (lean) and MetS pigs revealed that as compared with the control pigs, the MetS pigs had significantly more collagen in the muscularis layer, but not in the submucosa or mucosa layer. In conclusion, the Ossabaw pig model for diet-induced MetS is associated with oxidative stress and profibrotic activity in the bladder, which results in DU. This has previously been shown in mice and rats, but never in pigs. This novel model will better represent human MetS and DBD because the mechanism and size of the pig bladder more closely resemble that of a human, resulting in a more valid model and facilitating further study into the signaling mechanisms responsible for this impairment.
- Published
- 2020
41. Aging-associated skeletal muscle defects in HER2/Neu transgenic mammary tumor model
- Author
-
Trupti Trivedi, Peter A. Crooks, Teresa Zimmers, Narsimha Reddy Penthala, Jianguo Liu, Khalid S. Mohammad, Mayuri S. Prasad, Ruizhong Wang, Brijesh Kumar, Max Jacobsen, Theresa A. Guise, Harikrishna Nakshatri, George E. Sandusky, Calli Maguire, Poornima Bhat-Nakshatri, and Gabriela Ovalle
- Subjects
medicine.medical_specialty ,lcsh:Internal medicine ,Skeletal muscle ,MyoD ,HER2/neu ,Article ,Cachexia ,Breast cancer ,Internal medicine ,medicine ,Cytokines/chemokines ,Progenitor cell ,lcsh:RC31-1245 ,Mammary tumor ,biology ,Chemistry ,NF‐κB ,Cancer ,medicine.disease ,Functional limitations ,medicine.anatomical_structure ,Endocrinology ,biology.protein ,medicine.symptom ,Muscle contraction - Abstract
Background Loss of skeletal muscle volume and functional limitations are poor prognostic markers in breast cancer patients. Several molecular defects in skeletal muscle including reduced myoblast determination protein 1 (MyoD) levels and increased protein turn over due to enhanced proteosomal activity have been suggested as causes of skeletal muscle loss in cancer patients. However, it is unknown whether molecular defects in skeletal muscle are dependent on tumour aetiology. Methods We characterized functional and molecular defects of skeletal muscle in mouse mammary tumour virus (MMTV)‐Neu (Neu+) mice (n = 6–12), an animal model that represents HER2 + human breast cancer, and compared the results with well‐characterized luminal B breast cancer model MMTV‐PyMT (PyMT+). Functional studies such as grip strength, rotarod performance, and ex vivo muscle contraction were performed to measure the effects of cancer on skeletal muscle. Expression of muscle‐enriched genes and microRNAs as well as circulating cytokines/chemokines were measured. Because nuclear factor‐kappaB (NF‐κB) pathway plays a significant role in skeletal muscle defects, the ability of NF‐κB inhibitor dimethylaminoparthenolide (DMAPT) to reverse skeletal muscle defects was examined. Results Neu+ mice showed skeletal muscle defects similar to accelerated aging. Compared with age and sex‐matched wild type mice, Neu+ tumour‐bearing mice had lower grip strength (202 ± 6.9 vs. 179 ± 6.8 g grip force, P = 0.0069) and impaired rotarod performance (108 ± 12.1 vs. 30 ± 3.9 s, P
- Published
- 2020
42. c-Fms signaling mediates neurofibromatosis Type-1 osteoclast gain-in-functions.
- Author
-
Yongzheng He, Steven D Rhodes, Shi Chen, Xiaohua Wu, Jin Yuan, Xianlin Yang, Li Jiang, Xianqi Li, Naoyuki Takahashi, Mingjiang Xu, Khalid S Mohammad, Theresa A Guise, and Feng-Chun Yang
- Subjects
Medicine ,Science - Abstract
Skeletal abnormalities including osteoporosis and osteopenia occur frequently in both pediatric and adult neurofibromatosis type 1 (NF1) patients. NF1 (Nf1) haploinsufficient osteoclasts and osteoclast progenitors derived from both NF1 patients and Nf1(+/-) mice exhibit increased differentiation, migration, and bone resorptive capacity in vitro, mediated by hyperactivation of p21(Ras) in response to limiting concentrations of macrophage-colony stimulating factor (M-CSF). Here, we show that M-CSF binding to its receptor, c-Fms, results in increased c-Fms activation in Nf1(+/) (-) osteoclast progenitors, mediating multiple gain-in-functions through the downstream effectors Erk1/2 and p90RSK. PLX3397, a potent and selective c-Fms inhibitor, attenuated M-CSF mediated Nf1(+/-) osteoclast migration by 50%, adhesion by 70%, and pit formation by 60%. In vivo, we administered PLX3397 to Nf1(+/-) osteoporotic mice induced by ovariectomy (OVX) and evaluated changes in bone mass and skeletal architecture. We found that PLX3397 prevented bone loss in Nf1(+/-)-OVX mice by reducing osteoclast differentiation and bone resorptive activity in vivo. Collectively, these results implicate the M-CSF/c-Fms signaling axis as a critical pathway underlying the aberrant functioning of Nf1 haploinsufficient osteoclasts and may provide a potential therapeutic target for treating NF1 associated osteoporosis and osteopenia.
- Published
- 2012
- Full Text
- View/download PDF
43. The Role of TGF-β in Bone Metastases
- Author
-
Theresa A. Guise, Trupti Trivedi, Khalid S. Mohammad, and Gabriel M. Pagnotti
- Subjects
TGF-β therapeutic targets ,Epithelial-Mesenchymal Transition ,Angiogenesis ,transforming growth factor-β (TGF-β) ,Cell ,Bone Neoplasms ,Review ,Microbiology ,Biochemistry ,Bone and Bones ,Bone resorption ,Prostate cancer ,immune cells ,Immune system ,bone metastases ,Transforming Growth Factor beta ,medicine ,Humans ,programmed cell death ligand (PD-L1) ,check-point inhibitors ,Molecular Biology ,business.industry ,Cancer ,medicine.disease ,QR1-502 ,medicine.anatomical_structure ,Cancer cell ,Cancer research ,business ,bone resorption ,Transforming growth factor - Abstract
Complications associated with advanced cancer are a major clinical challenge and, if associated with bone metastases, worsen the prognosis and compromise the survival of the patients. Breast and prostate cancer cells exhibit a high propensity to metastasize to bone. The bone microenvironment is unique, providing fertile soil for cancer cell propagation, while mineralized bone matrices store potent growth factors and cytokines. Biologically active transforming growth factor β (TGF-β), one of the most abundant growth factors, is released following tumor-induced osteoclastic bone resorption. TGF-β promotes tumor cell secretion of factors that accelerate bone loss and fuel tumor cells to colonize. Thus, TGF-β is critical for driving the feed-forward vicious cycle of tumor growth in bone. Further, TGF-β promotes epithelial-mesenchymal transition (EMT), increasing cell invasiveness, angiogenesis, and metastatic progression. Emerging evidence shows TGF-β suppresses immune responses, enabling opportunistic cancer cells to escape immune checkpoints and promote bone metastases. Blocking TGF-β signaling pathways could disrupt the vicious cycle, revert EMT, and enhance immune response. However, TGF-β’s dual role as both tumor suppressor and enhancer presents a significant challenge in developing therapeutics that target TGF-β signaling. This review presents TGF-β’s role in cancer progression and bone metastases, while highlighting current perspectives on the therapeutic potential of targeting TGF-β pathways.
- Published
- 2021
- Full Text
- View/download PDF
44. Neuropeptide Y regulates a vascular gateway for hematopoietic stem and progenitor cells
- Author
-
Malgorzata M. Kamocka, Khalid S. Mohammad, Nadia Carlesso, Jonathan Hoggatt, Mary R. Saunders, Louis M. Pelus, Pratibha Singh, Theresa A. Guise, Jennifer M. Speth, and Hongge Li
- Subjects
0301 basic medicine ,Dipeptidyl Peptidase 4 ,Vascular permeability ,Mice ,03 medical and health sciences ,mental disorders ,Animals ,Humans ,Neuropeptide Y ,Progenitor cell ,Receptor ,Mice, Knockout ,Chemistry ,Endothelial Cells ,General Medicine ,Hematopoietic Stem Cells ,Neuropeptide Y receptor ,humanities ,Receptors, Neuropeptide Y ,Cell biology ,Endothelial stem cell ,Transplantation ,Haematopoiesis ,030104 developmental biology ,Commentary ,Stem cell ,Signal Transduction - Abstract
Endothelial cells (ECs) are components of the hematopoietic microenvironment and regulate hematopoietic stem and progenitor cell (HSPC) homeostasis. Cytokine treatments that cause HSPC trafficking to peripheral blood are associated with an increase in dipeptidylpeptidase 4/CD26 (DPP4/CD26), an enzyme that truncates the neurotransmitter neuropeptide Y (NPY). Here, we show that enzymatically altered NPY signaling in ECs caused reduced VE-cadherin and CD31 expression along EC junctions, resulting in increased vascular permeability and HSPC egress. Moreover, selective NPY2 and NPY5 receptor antagonists restored vascular integrity and limited HSPC mobilization, demonstrating that the enzymatically controlled vascular gateway specifically opens by cleavage of NPY by CD26 signaling via NPY2 and NPY5 receptors. Mice lacking CD26 or NPY exhibited impaired HSPC trafficking that was restored by treatment with truncated NPY. Thus, our results point to ECs as gatekeepers of HSPC trafficking and identify a CD26-mediated NPY axis that has potential as a pharmacologic target to regulate hematopoietic trafficking in homeostatic and stress conditions.
- Published
- 2017
- Full Text
- View/download PDF
45. Halofuginone inhibits TGF-β/BMP signaling and in combination with zoledronic acid enhances inhibition of breast cancer bone metastasis
- Author
-
Khalid S. Mohammad, Pierrick G.J. Fournier, John M. Chirgwin, Holly W. Davis, Xiang H. Peng, Alain Mauviel, Ryan C. McKenna, Theresa A. Guise, Patricia Juárez, and Maria Niewolna
- Subjects
0301 basic medicine ,TGF-β ,Pathology ,medicine.medical_specialty ,Osteolysis ,03 medical and health sciences ,Prostate cancer ,zoledronic acid ,0302 clinical medicine ,Breast cancer ,halofuginone ,bone metastases ,medicine ,BMP ,Halofuginone ,business.industry ,Bone metastasis ,medicine.disease ,3. Good health ,030104 developmental biology ,Zoledronic acid ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,business ,Transforming growth factor ,medicine.drug ,Research Paper - Abstract
// Patricia Juarez 1, 2 , Pierrick G.J. Fournier 1, 2 , Khalid S. Mohammad 1 , Ryan C. McKenna 3 , Holly W. Davis 3 , Xiang H. Peng 1 , Maria Niewolna 1 , Alain Mauviel 4, 5, 6, 7 John M. Chirgwin 1 and Theresa A. Guise 1 1 Division of Endocrinology, Department of Medicine, Indiana University Purdue University at Indianapolis, Indiana, USA 2 Ensenada Center for Scientific Research and Higher Education, Ensenada, Mexico 3 University of Virginia, Charlottesville, Virginia, USA 4 Institute Curie, Orsay, France 5 INSERM U1021, Orsay, France 6 CNRS UMR3347, Orsay, France 7 Universite Paris XI, Orsay, France Correspondence to: Theresa A. Guise, email: tguise@iu.edu Keywords: halofuginone, zoledronic acid, bone metastases, TGF-β, BMP Received: June 21, 2017 Accepted: August 04, 2017 Published: September 23, 2017 ABSTRACT More efficient therapies that target multiple molecular mechanisms are needed for the treatment of incurable bone metastases. Halofuginone is a plant alkaloid-derivative with antiangiogenic and antiproliferative effects. Here we demonstrate that halofuginone is an effective therapy for the treatment of bone metastases, through multiple actions that include inhibition of TGFβ and BMP-signaling. Halofuginone blocked TGF-β-signaling in MDA-MB-231 and PC3 cells showed by inhibition of TGF-β-induced Smad-reporter, phosphorylation of Smad-proteins, and expression of TGF-β-regulated metastatic genes. Halofuginone increased inhibitory Smad7-mRNA and reduced TGF-β-receptor II protein. Proline supplementation but not Smad7-knockdown reversed halofuginone-inhibition of TGF-β-signaling. Halofuginone also decreased BMP-signaling. Treatment of MDA-MB-231 and PC3 cells with halofuginone reduced the BMP-Smad-reporter (BRE) 4 , Smad1/5/8-phosphorylation and mRNA of the BMP-regulated gene Id-1. Halofuginone decreased immunostaining of phospho-Smad2/3 and phospho-Smad1/5/8 in cancer cells in vivo . Furthermore, halofuginone decreased tumor-take and growth of orthotopic-tumors. Mice with breast or prostate bone metastases treated with halofuginone had significantly less osteolysis than control mice. Combined treatment with halofuginone and zoledronic-acid significantly reduced osteolytic area more than either treatment alone. Thus, halofuginone reduces breast and prostate cancer bone metastases in mice and combined with treatment currently approved by the FDA is an effective treatment for this devastating complication of breast and prostate-cancer.
- Published
- 2017
46. Improved autologous cortical bone harvest and viability with 2Flute otologic burs
- Author
-
Khalid S. Mohammad, Michael J. Ye, Pei-Ciao Tang, Adam A. Roth, and Rick F. Nelson
- Subjects
medicine.medical_specialty ,biology ,business.industry ,medicine.medical_treatment ,Cholesteatoma ,Dentistry ,Osteoblast ,Mastoidectomy ,030206 dentistry ,Bone healing ,medicine.disease ,Surgery ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Otorhinolaryngology ,medicine ,Osteocalcin ,biology.protein ,Alkaline phosphatase ,Cortical bone ,Tympanomastoidectomy ,business ,030217 neurology & neurosurgery - Abstract
Objectives To determine if 2Flute (Stryker Corporation, Kalamazoo, MI) otologic burs improve the size, cellular content, and bone healing of autologous cortical bone grafts harvested during canal wall reconstruction (CWR) tympanomastoidectomy with mastoid obliteration. Study Design Institutional review board-approved prospective cohort study. Methods Human autologous cortical bone chips were harvested using various burs (4 and 6 mm diameter; multiflute, and 2Flute [Stryker Corporation]) from patients undergoing CWR tympanomastoidectomy for the treatment of chronic otitis media with cholesteatoma. Bone chip size, cell counts, cellular gene expression, and new bone formation were quantified. Results Bone chips were significantly larger when harvested with 2Flute (Stryker Corporation) bur compared to multiflute burs at both 6 mm diameter (113 ± 14 μm2 vs. 66 ± 8 μm2; P
- Published
- 2017
- Full Text
- View/download PDF
47. The vitamin D receptor is involved in the regulation of human breast cancer cell growth via a ligand-independent function in cytoplasm
- Author
-
Sutha John, Suzanne Schillo, Colin R. Dunstan, Hong Zhou, Yu Zheng, Theresa A. Guise, Pierrick G.J. Fournier, Khalid S. Mohammad, Sreemala Murthy, Trupti Trivedi, and Markus J. Seibel
- Subjects
0301 basic medicine ,Cytoplasm ,Gene Expression ,Apoptosis ,vitamin D ,Ligands ,Calcitriol receptor ,Mice ,0302 clinical medicine ,polycyclic compounds ,bone metastasis ,Gene knockdown ,digestive, oral, and skin physiology ,Bone metastasis ,3. Good health ,Protein Transport ,Oncology ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Heterografts ,lipids (amino acids, peptides, and proteins) ,Female ,Signal transduction ,Osteosclerosis ,Research Paper ,musculoskeletal diseases ,medicine.medical_specialty ,ligand independent ,Bone Neoplasms ,Breast Neoplasms ,03 medical and health sciences ,breast cancer ,Cell Line, Tumor ,Internal medicine ,medicine ,Vitamin D and neurology ,Animals ,Humans ,vitamin D receptor ,Cell Proliferation ,business.industry ,Cell growth ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Mutation ,Cancer cell ,Cancer research ,Receptors, Calcitriol ,business - Abstract
// Trupti Trivedi 1, 2 , Yu Zheng 1 , Pierrick G.J. Fournier 2, 5 , Sreemala Murthy 2 , Sutha John 2 , Suzanne Schillo 1 , Colin R. Dunstan 3 , Khalid S. Mohammad 2 , Hong Zhou 1 , Markus J. Seibel 1, 4 , Theresa A. Guise 2 1 Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia 2 Division of Endocrinology, Department of Medicine, Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana, USA 3 Department of Biomedical Engineering, University of Sydney, Sydney, Australia 4 Department of Endocrinology and Metabolism, Concord Hospital, Concord, Sydney, Australia 5 Biomedical Innovation Department, Scientific Research and High Education Center from Ensenada (CICESE), Ensenada, Baja California, Mexico Correspondence to: Theresa A. Guise, email: tguise@iu.edu Markus J. Seibel, email: markus.seibel@sydney.edu.au Keywords: breast cancer, vitamin D, vitamin D receptor, ligand independent, bone metastasis Received: November 04, 2016 Accepted: February 15, 2017 Published: March 01, 2017 ABSTRACT Vitamin D has pleiotropic effects on multiple tissues, including malignant tumors. Vitamin D inhibits breast cancer growth through activation of the vitamin D receptor (VDR) and via classical nuclear signaling pathways. Here, we demonstrate that the VDR can also function in the absence of its ligand to control behaviour of human breast cancer cells both outside and within the bone microenvironment. Stable shRNA expression was used to knock down VDR expression in MCF-7 cells, generating two VDR knockdown clonal lines. In ligand-free culture, knockdown of VDR in MCF-7 cells significantly reduced proliferation and increased apoptosis, suggesting that the VDR plays a ligand-independent role in cancer cell growth. Implantation of these VDR knockdown cells into the mammary fat pad of nude mice resulted in reduced tumor growth in vivo compared with controls. In the intra-tibial xenograft model, VDR knockdown greatly reduced the ability of the cells to form tumors in the bone microenvironment. The in vitro growth of VDR knockdown cells was rescued by the expression of a mutant form of VDR which is unable to translocate to the nucleus and hence accumulates in the cytoplasm. Thus, our data indicate that in the absence of ligand, the VDR promotes breast cancer growth both in vitro and in vivo and that cytoplasmic accumulation of VDR is sufficient to produce this effect in vitro . This new mechanism of VDR action in breast cancer cells contrasts the known anti-proliferative nuclear actions of the VDR-vitamin D ligand complex.
- Published
- 2017
- Full Text
- View/download PDF
48. The P2Y 2 nucleotide receptor is an inhibitor of vascular calcification
- Author
-
Jenna N. Regan, April M. Hoggatt, Cheikh I. Seye, Maxwell T. Shelton, Paul Herring, Khalid S. Mohammad, and Shaomin Qian
- Subjects
0301 basic medicine ,Apolipoprotein E ,medicine.medical_specialty ,Vascular smooth muscle ,Apyrase ,Biology ,medicine.disease ,03 medical and health sciences ,NT5E ,Arterial calcification ,030104 developmental biology ,Endocrinology ,Internal medicine ,Nucleotidase ,medicine ,Receptor ,Cardiology and Cardiovascular Medicine ,Calcification - Abstract
Background and aims Mutations in the 5'-nucleotidase ecto (NT5E) gene that encodes CD73, a nucleotidase that converts AMP to adenosine, are linked to arterial calcification. However, the role of purinergic receptor signaling in the pathology of intimal calcification is not well understood. In this study, we examined whether extracellular nucleotides acting via P2Y 2 receptor (P2Y 2 R) modulate arterial intimal calcification, a condition highly correlated with cardiovascular morbidity. Methods Apolipoprotein E, P2Y 2 R double knockout mice ( ApoE −/− P2Y 2 R −/− ) were used to determine the effect of P2Y 2 R deficiency on vascular calcification in vivo . Vascular smooth muscle cells (VSMC) isolated from P2Y 2 R −/− mice grown in high phosphate medium were used to assess the role of P2Y 2 R in the conversion of VSMC into osteoblasts. Luciferase-reporter assays were used to assess the effect of P2Y 2 R on the transcriptional activity of Runx2. Results P2Y 2 R deficiency in ApoE −/− mice caused extensive intimal calcification despite a significant reduction in atherosclerosis and macrophage plaque content. The ectoenzyme apyrase that degrades nucleoside di- and triphosphates accelerated high phosphate-induced calcium deposition in cultured VSMC. Expression of P2Y 2 R inhibits calcification in vitro inhibited the osteoblastic trans -differentiation of VSMC. Mechanistically, expression of P2Y 2 R inhibited Runx2 transcriptional activation of an osteocalcin promoter driven luciferase reporter gene. Conclusions This study reveals a role for vascular P2Y 2 R as an inhibitor of arterial intimal calcification and provides a new mechanistic insight into the regulation of the osteoblastic trans -differentiation of SMC through P2Y 2 R-mediated Runx2 antagonism. Given that calcification of atherosclerotic lesions is a significant clinical problem, activating P2Y 2 R may be an effective therapeutic approach for treatment or prevention of vascular calcification.
- Published
- 2017
- Full Text
- View/download PDF
49. A Single Radioprotective Dose of Prostaglandin E
- Author
-
Andrea M, Patterson, Liqiong, Liu, Carol H, Sampson, P Artur, Plett, Hongge, Li, Pratibha, Singh, Khalid S, Mohammad, Jonathan, Hoggatt, Maegan L, Capitano, Christie M, Orschell, and Louis M, Pelus
- Subjects
p53 ,hematopoietic regeneration ,bone marrow ,Transcription, Genetic ,Apoptosis ,Radiation-Protective Agents ,Dinoprostone ,Article ,stem cells ,Radiation, Ionizing ,Animals ,Gene Regulatory Networks ,16,16-dimethyl PGE2 ,Cell Cycle ,RNA sequencing ,Hematopoietic Stem Cells ,Hematopoiesis ,Mice, Inbred C57BL ,radiation ,Gene Expression Regulation ,DNA damage ,cell cycle ,prostaglandin ,DNA Damage ,Signal Transduction ,Transcription Factors - Abstract
Summary Ionizing radiation exposure results in acute and delayed bone marrow suppression. Treatment of mice with 16,16-dimethyl prostaglandin E2 (dmPGE2) prior to lethal ionizing radiation (IR) facilitates survival, but the cellular and molecular mechanisms are unclear. In this study we show that dmPGE2 attenuates loss and enhances recovery of bone marrow cellularity, corresponding to a less severe hematopoietic stem cell nadir, and significantly preserves long-term repopulation capacity and progenitor cell function. Mechanistically, dmPGE2 suppressed hematopoietic stem cell (HSC) proliferation through 24 h post IR, which correlated with fewer DNA double-strand breaks and attenuation of apoptosis, mitochondrial compromise, oxidative stress, and senescence. RNA sequencing of HSCs at 1 h and 24 h post IR identified a predominant interference with IR-induced p53-downstream gene expression at 1 h, and confirmed the suppression of IR-induced cell-cycle genes at 24 h. These data identify mechanisms of dmPGE2 radioprotection and its potential role as a medical countermeasure against radiation exposure., Graphical Abstract, Highlights • Treatment with dmPGE2 prior to lethal IR attenuates early hematopoietic cell loss • IR-induced apoptotic gene expression is blocked in HSCs by dmPGE2 • dmPGE2 temporarily suppresses proliferation of HSCs and HPCs in vivo • Early IR-induced cycling, DNA damage, and their sequelae were attenuated in HSCs, In this article, Pelus, Orschell, and colleagues demonstrate that dmPGE2 protection from lethal radiation preserves HSC numbers and repopulating capacity, which is associated with attenuation of HSC cycling and DNA damage accumulation within the first critical day. Genomically, dmPGE2 predominantly blocks HSC induction of p53-regulated apoptotic genes within 1 h of irradiation, altogether promoting hematopoietic recovery and survival.
- Published
- 2020
50. Mechanical suppression of breast cancer cell invasion and paracrine signaling to osteoclasts requires nucleo-cytoskeletal connectivity
- Author
-
Gabriel M. Pagnotti, Xin Yi, Gunes Uzer, William R. Thompson, Uma Sankar, Khalid S. Mohammad, Theresa A. Guise, Joseph M. Wallace, Laura E. Wright, Katherine M Powell, and Clinton T. Rubin
- Subjects
Gene knockdown ,Histology ,lcsh:QP1-981 ,biology ,Physiology ,Chemistry ,Endocrinology, Diabetes and Metabolism ,LINC complex ,Cancer ,medicine.disease ,Article ,lcsh:Physiology ,Bone quality and biomechanics ,Cell biology ,Paracrine signalling ,medicine.anatomical_structure ,lcsh:Biology (General) ,Osteoclast ,RANKL ,Cancer cell ,Bone cancer ,medicine ,biology.protein ,Secretion ,lcsh:QH301-705.5 - Abstract
Exercise benefits the musculoskeletal system and reduces the effects of cancer. The effects of exercise are multifactorial, where metabolic changes and tissue adaptation influence outcomes. Mechanical signals, a principal component of exercise, are anabolic to the musculoskeletal system and restrict cancer progression. We examined the mechanisms through which cancer cells sense and respond to low-magnitude mechanical signals introduced in the form of vibration. Low-magnitude, high-frequency vibration was applied to human breast cancer cells in the form of low-intensity vibration (LIV). LIV decreased matrix invasion and impaired secretion of osteolytic factors PTHLH, IL-11, and RANKL. Furthermore, paracrine signals from mechanically stimulated cancer cells, reduced osteoclast differentiation and resorptive capacity. Disconnecting the nucleus by knockdown of SUN1 and SUN2 impaired LIV-mediated suppression of invasion and osteolytic factor secretion. LIV increased cell stiffness; an effect dependent on the LINC complex. These data show that mechanical vibration reduces the metastatic potential of human breast cancer cells, where the nucleus serves as a mechanosensory apparatus to alter cell structure and intercellular signaling.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.