Dr. Ahmed Yaqinuddin is a distinguished researcher in the field of epigenetics, with a particular emphasis on the epigenetic mechanisms that underlie a variety of chronic diseases and malignancies, such as genitourinary, liver, and gastrointestinal (GI) cancers. His extensive research focuses on the complex mechanisms by which epigenetic modifications, including DNA methylation and microRNA expression, contribute to the development and progression of these malignancies. Dr. Yaqinuddin's research endeavors to establish a connection between laboratory discoveries and clinical applications. He is profoundly committed to the advancement of point-of-care diagnostic tests that can aid in the early detection of cancer. Dr. Yaqinuddin's innovative approaches have the potential to revolutionize cancer screening and management by providing minimally invasive, efficient, and accurate diagnostic tools that are based on cell-free microRNAs and DNA methylation markers found in body fluids.
Dr. Yaqinuddin has made significant contributions to the comprehension of the inflammatory processes that are involved in a variety of diseases in addition to his cancer research. His research has illuminated the role of Neutrophil Extracellular Traps (NETs) in the pathophysiology of COVID-19, demonstrating how NETs contribute to severe disease manifestations and complications. He has broadened this investigation to encompass chronic conditions, including Alzheimer's disease, diabetes, and a variety of malignancies, thereby clarifying the intricate relationship between inflammation, immunity, and the development of chronic diseases.
Dr. Yaqinuddin's interdisciplinary approach integrates clinical research, epigenetics, and molecular biology to confront some of the most urgent issues in contemporary medicine. His pioneering research has paved the way for significant progress in the early diagnosis and treatment of cancer, as well as a comprehensive understanding of chronic diseases related to inflammation.
His research interests include translational research, molecular characterization of cancers and therapeutic targets. He recently focused on high throughput sequencing techniques for analyzing mutations in different cancers such as thyroid, colorectal, and breast. Targeting these identified molecular and genetic alterations using either molecular inhibitors, small interference RNA (siRNA) or antibodies are also performed to identify genes that can be therapeutically targeted for the improvement of cancer therapy.
The role of cancer-associated fibroblasts in breast carcinogenesis and the role of secreted cytokines in this crosstalk between cancer cells and their protective fibroblasts. We are aslo interested in developing drugs to target both cancer cells and also their associated fibroblasts. These drugs are also tested against other types of cancer such as ovarian, colerectal, and bone.
Scopus Name: Islam M. SaadeldinEmail: imohamed@kfshrc.edu.sa
Dr. Saadeldin's research extends across various facets of reproductive biotechnology, including somatic cell nuclear transfer (SCNT), transgenesis, endometrial organoids, adult, and embryonic stem cells. Dr. Saadeldin's scholarly pursuits delve into the intricate roles played by extracellular vesicles or exosomes in embryo communication and modeling the dynamic interactions between embryonic and maternal compartments.
One of his notable research directions involves studying comparative cellular defense mechanisms against extreme hyperthermia, exploring correlations with cellular anastasis and resilience. Currently, his research trajectory is centered around the development of CRISPR/Cas9-engineered extracellular vesicles, with the overarching goal of enhancing embryo implantation and improving pregnancy outcomes. Additionally, he is leading a project regarding the oncofertility and how to preserve the female fertility from the chemotherapy through the use of stem cells-derived exosomes.
Currently, Dr. Alhamlan’s lab is spearheading the Microbial Genomic and Epidemiological Surveillance System at KFSH&RC. Research focuses mainly on emerging and re-emerging pathogens, sexually transmitted infections (STIs), HPV-related cancers, and microbiomes. Additionally, we are actively engaged in developing in-house diagnostic tests, including localized encephalitis and CMV drug-resistance tests, to ensure prompt and accurate diagnoses for ultimate patient care.
Breast cancer is the most common cancer in women worldwide. Despite progress in drug development against specific target in breast cancer, tumor related mortality remain high due disease relapse. Fascin is an actin-bundling protein that get expressed in many type of cancers including breast. Impotently, our lab demonstrated that fascin expression levels is linked to many poor prognostic markers including drug resistance and metastasis, major contributors in disease relapse and shorter survival. Breast cancer are classified into 4 major molecular subtypes. Luminal A), luminal B, HER2-enriched and triple negative breast cancer (TNBC). Subtyping of breast cancer is required for defining the appropriate treatment and distinguishing the good responders from high risk subtype. Fascin is expressed in 15-25% of luminal A and B, which account for around 70% of all cases and are the least aggressive of the 4 breast cancer subtypes. HER2-encirhced (40-50% are fascin positive) accounts for 20-30% of diagnosed breast cancers and more aggressive than luminal, but is less aggressive than TNBC (85% are fascin positive) which makes up between 15-20% of the cases. Luminal A and B breast cancer have better prognosis due to the high sensitivity to anti-estrogen hormonal therapy, while HER2-enirched breast cancer can be treated with anti-HER2 antibodies, adjuvant and/or chemotherapy. However, TNBC are the most aggressive and had the least therapeutic options. However, disease relapse remain high among breast cancer patients, thus necessitating the need for an alternative therapy. Since we established a direct link between fascin expression and poor prognosis, our lab is focused on manipulating fascin expression in different breast cancer cell line model. This help us to combines in vitro assay, preclinical studies in animal and patient sample as a first step toward understanding fascin’s function in the progression of breast cancer. Ongoing clinical trials using small molecule targeting fascin in solo or in combination with other therapies showed promising results for multiple type of malignancies. Deep understating of fascin’s function in breast cancer cells may help in designing the best combinational therapy especially for drug resistant and metastatic breast cancer.
Nanostructure Materials for Biomedical application including theragnostic platforms, drug delivery systems, biomaterial/device coatings, tissue engineering scaffolds, performance-enhanced therapeutic alternatives.
Prof. Namik Kaya (earned his PhD in 1997, The Ohio State University, Columbus, OH, USA). Currently, he is a senior scientist and head of Neurogenetics Unit in Translational Genomics department at the Center for Genomic Medicine, KFSHRC. He received his professorship in human genetics/genomics in 2018.
Dr. Namik Kaya has received his PhD from the Ohio State University in 1997. He did his postdoctoral studies at the same university. He joined King Faisal Specialist Hospital and Research Centre (KFSHRC) in 2004. He then established Neurogenetics Unit within the Department of Genetics. Dr. Kaya’s research interests include genetics of neurological disorders, particularly ataxias, leukodystrophies, and channelopathies, genetics of neurodevelopmental disorders, mitochondrial disorders, and integrated omics for precision medicine. He has a strong track record of scientific publication and securing grant funds. Currently he is principal investigator of several critical projects (including an international clinical trial) several of which are funded by different funding agencies. He has over 100 scientific publications in various peer-reviewed international journals. His lab discovered several disease-causing novel genes and novel syndromes such as KAYA-BARAKAT-MASSON Syndrome, OMIM# 619125; and BARAKAT-VAN HAM-KAYA Syndrome, OMIM# 619389. Dr. Kaya holds Professorship in Human Genetics and Genomics and was the recipient of KFSHRC’s prestigious “The Best Publication Award” for six consecutive years. He has been widely recognized for his training and educational activities and received numerous recognition awards for his efforts. Dr. Kaya has been serving on the editorial board (Genetics Section) of PLoS ONE, and reviews grants for funding agencies such as King Abdulaziz City of Science and Technology and King Salman Center for Disability Research. He is member of American Society of Human Genetics (ASHG), European Society of Human Genetics, and Society for Neuroscience. Currently, he is leading Neurogenetics Unit in Translational Genomics Department at the Center for Genomic Medicine, KFSHRC.
Associate professor of Cancer Biology
My laboratory investigates signaling pathways that control apoptosis in cancer cells. We identified PI3K/Akt as dominant anti-apoptotic pathway activated by IGF-1 in fibroblasts and by EGF in epithelial cells. Subsequently, we have described a network of signaling pathways that regulate survival of prostate cancer cells and identified BAD (a BH3-only member of Bcl2 family) as a downstream convergence node of multiple anti-apoptotic signaling pathways ( PI3K/Akt, EGFR and GPCRs). We were first to show that stress hormone epinephrine protects prostate cancer cells from apoptosis via PKA/BAD signaling and demonstrated that Epi/ ADRB2/PKA/BAD accelerates progression of prostate tumors in mice.
I have directed multi-investigator research project that demonstrated the role of ADRB2 signaling in prostate cancer and collaborative projects on the development of prostate-selective PI3K inhibitors and prostate-targeted toxins.
Currently my interests shifted to the analysis of signaling pathways that operate in normal prostate and large intestine and in malignancies of these tissues.
Evidence-based medicine and evidence-based statistics. The practical application of evidential statistics (likelihood approach) to the analysis of healthcare data. The in vitro neuropharmacological study of Merkel cell mechanoreceptors.
Professor in Medical Biochemistry
My research focuses on two key areas within the fields of molecular medicine and pharmacogenomics:
Metformin/Sulfonylurea Combination Therapy 6-Month Follow-Up Study. Journal of Personalized Medicine.2020 (10), 53-61.
Know More.Dr. Khanfar’s research involves the use of state-of-the-art pharmaceutical, computational, and medicinal chemistry technologies to design, synthesize, and evaluate new molecules for the treatment of human diseases and to probe biological systems, with a particular emphasis on cancer and neurodegenerative diseases. His research is pioneering in the development of novel computational and molecular modeling technologies aimed at understanding protein-drug interactions and discovering novel, active drugs.
Dr. Khanfar’s research team employs a comprehensive suite of computer-aided drug design methods. These include multidimensional quantitative structure-activity relationship (QSAR) models, which correlate the chemical structure of compounds with their biological activity, docking techniques that predict the preferred orientation of one molecule to a second when bound to each other, conformational analysis to understand the shape and flexibility of molecules, and pharmacophore modeling to identify the structural features responsible for a drug's biological activity. These methods collectively enhance the understanding of drug action and facilitate the design and discovery of new therapeutics.
In addition to computational techniques, Dr. Khanfar applies synthetic and medicinal chemistry methods to design and synthesize new therapeutics, specifically targeting oncology and neurodegenerative diseases. His innovative work in synthetic chemistry has led to the creation of several chemical scaffolds aimed at cancer-related kinases. These kinases play crucial roles in the regulation of cell functions, and their dysregulation is often associated with cancer. Moreover, Dr. Khanfar has developed modulators of Histamine subtype 3 (H3 receptors), which are significant in the context of neurodegenerative diseases such as Parkinson's and Alzheimer's diseases. These modulators have the potential to improve cognitive functions and offer new therapeutic avenues for treating these debilitating conditions.
My research primarily focuses on the complex interactions between cancer and bone. Specifically, I am dedicated to investigating how the microenvironment of the bone influences the behavior of tumors and their response to various therapeutic interventions. My focus lies on deciphering the molecular mechanisms that underlie the development of bone metastases and the systemic affects they entail. An integral part of my research is looking into the complex role played by transforming growth factor-beta (TGF-β) in the normal remodeling processes of bone and the progression of metastatic lesions. I am particularly interested in elucidating how TGF-β modulates the activities of osteoblasts and osteoclasts, influencing the advancement of cancer within the bone microenvironment.
Another area of exploration within my research pertains to investigating the contributions of bone-derived TGF-β and other signaling molecules in regulating glucose metabolism. I focus on the involvement of bone marrow adipocytes in the context of bone metastases, offering valuable insights into the metabolic dysfunctions associated with cancer progression.
My research efforts extend to the examination of the influence of endothelin-1 on bone remodeling processes and the formation of osteoblastic metastases. Through a detailed analysis of the signaling pathways activated by endothelin-1 that promote osteoblastic activity and bone formation, I aim to identify new therapeutic targets that could revolutionize the treatment of bone metastases. I am actively involved in studying the impacts of anti-VEGF therapies on both bone metastases and normal bone development, with the goal of refining cancer treatment strategies to minimize adverse effects on bone health.
Another aspect of my research involves the rigorous evaluation of novel anticancer compounds using animal models of bone metastases. This preclinical research phase is indispensable for assessing the efficacy and safety profiles of potential therapeutic agents before they can progress to the clinical trial stage.
Prof. Zourob leading the Biosensors BioMEMS and Bionanotechnology Lab (BBBL) at Alfaisal University. The lab has more than 40 active researchers including graduate students, undergraduate students, postdoctoral and visitors from different parts of the world. The lab is a multidisciplinary, where the lab members coming from different backgrounds including science, nanotechnology, biomedical science, pharmacy, medicine, radiology, and engineering. BBBL is extensively engaged with local and international collaborations. More than 120 graduate students and postdoctoral scientists already trained/graduated from the BBBL lab and secured positions in various academic institutions, industry, government jobs, and research entities locally and internationally. The lab is very active in developing rapid diagnostic/detection tools, specific recognition elements, lab on a chip for sample processing and organ on a chip. The lab has active research in developing low-cost paper, colorimetric, electrochemical, optical and soft and flexible materials based-diagnostic tools for various biomedical applications. Another axis of research at BBBL includes the development of various recognition elements such as immunoassays, aptamers, peptides and various synthetic recognition elements. Normally, Prof. Zourob discusses with the potential students who is interested to join the lab, their research interest, technical skills, future plans and try to design a research project which fits with both parties’ interest. Therefore, please contact him and discuss your interest. Nothing to lose.
Motivated by an unyielding enthusiasm for unraveling the cellular processes that form the foundation of health and disease, my commitment lies in advancing the field of research pertaining to the mechanisms governing metabolic disorders and cancer. At the core of my investigations lies the phenomenon known as stress-induced premature senescence (SIPS), which plays a pivotal role in the emergence and progression of these debilitating ailments. My objective is to unveil the molecular basis of SIPS, identify novel biomarkers, and pave the way for therapeutic strategies. SIPS manifests through the emergence of senescence-associated traits, such as growth arrest and heightened expression of specific markers, in cells that have been exposed to stress. It is implicated in a multitude of diseases, including cancer, cardiovascular disease, and metabolic disorders. The mechanisms that underlie SIPS involve a complex interplay of factors, including telomere dysfunction, DNA damage, epigenetic alterations, and mitochondrial dysfunction. Cellular stressors have the capacity to inflict DNA damage, thereby prompting cells to initiate repair mechanisms and subsequently enter a state of growth arrest, ultimately leading to SIPS. Epigenetic modifications, such as DNA methylation, also contribute to the induction of SIPS. Furthermore, mitochondrial dysfunction, which impairs the cell's ability to generate energy, contributes to the onset of SIPS. Given the nature of SIPS and its association with a diverse range of diseases, further research is imperative in order to comprehensively comprehend its mechanisms and develop innovative therapeutic approaches to prevent and treat pathologies related to SIPS. My research employs cutting-edge methodologies, including metabolomics, proteomics, lipidomics, and transcriptomics, to delve into these areas.
Current Research Activities: My research interests encompasses experimental oncology and diagnostic pathology are directed at Translational Clinical Proteomics and biomarker discovery for clinical applications to different diseases in general. My mission of translational research towards Personalized Medicine mainly focus on molecular analysis of clinical samples from patients; including epithelial solid tumors, hematological malignancies, other non-malignant diseases, bodily fluids, stem cell subtypes as well as experimental cell types using Expression Proteomics approach. My life science research expertise is directed in mapping the molecular alterations involved in different disease conditions using state-of the-art proteomics technologies. Currently studying the complex protein expression patterns that may be decisive for the accurate diagnosis, biological behavior, treatment sensitivity and prognosis of specific human tumors. Others are biomarker discovery for kidney diseases and annotation of proteome maps for some selected human derived stem cells. I have well over 65 publications with in the field of Medical Proteomics in high impact factor peer reviewed journals. My research work have convincingly demonstrated Proof of concept studies that proteome analysis can be further developed into a “proteomedical scanner”, i.e., an artificial intelligence tool (protein fingerprint barcode) capable of assisting clinical decision makings in establishing a more accurate diagnosis and prognosis. With my proteomics research initiatives expertise, I have established international collaborations with academic institutions in Europe as well the Gulf region including King Faisal Specialist Hospital and Research Center Riyadh, Alfaisal University, Riyadh, KFMC, national Guard Hospital, University of Bahrain, Karolinska Institute and engaged in basic and translational clinical research and training and supervision of graduate students. I have years of experience of working with Institution review board including as member of Clinical Research Committee, Research ethics committee, and Basic Research Committee. I am keen to develop new innovative projects and collaborations with national and international research groups hopefully, generating new information and testing the real value of the technology in clinical practice.
My interest is mainly in reproductive physiology and infertility. At the beginning of my carrier, I involved in intensive research in vitro oocyte maturation and effect of growth factors on the process in animals and human gametes. Currently, most of my research is in genetics of infertility and preimplanatation genetic testing. Moreover, morphokinetics of early embryo development and its relation to embryo development and treatment success by using artificial intelligence model are in my current research projects.
My past research activity has been focused mainly on viral immunopathology with respect to cytokines, T cell and macrophage activity, electron microscopy and bioinformatics. In recent years, I have been involved more with looking at the antibacterial effects of nanoparticles and the cytotoxic effects of various natural compounds.
Dr. Baltatu's research focuses on translational medicine and biomarker discovery, particularly in the areas of: