Catalogue Search | MBRL
Search Results Heading
Explore the vast range of titles available.
MBRLSearchResults
-
DisciplineDiscipline
-
Is Peer ReviewedIs Peer Reviewed
-
Item TypeItem Type
-
SubjectSubject
-
YearFrom:-To:
-
More FiltersMore FiltersSourceLanguage
Done
Filters
Reset
50
result(s) for
"Golmohammadi, Maryam"
Sort by:
Role of interferon therapy in severe COVID-19: the COVIFERON randomized controlled trial
by
Mokhtari, Majid
,
Amirdosara, Mahdi
,
Zali, Alireza
in
692/699/1785
,
692/699/255
,
Adverse events
2021
Type 1 Interferons (IFNs) have been associated with positive effects on Coronaviruses. Previous studies point towards the superior potency of IFNβ compared to IFNα against viral infections. We conducted a three-armed, individually-randomized, open-label, controlled trial of IFNβ1a and IFNβ1b, comparing them against each other and a control group. Patients were randomly assigned in a 1:1:1 ratio to IFNβ1a (subcutaneous injections of 12,000 IU on days 1, 3, 6), IFNβ1b (subcutaneous injections of 8,000,000 IU on days 1, 3, 6), or the control group. All three arms orally received Lopinavir/Ritonavir (400 mg/100 mg twice a day for ten days) and a single dose of Hydroxychloroquine 400 mg on the first day. Our utilized primary outcome measure was Time To Clinical Improvement (TTCI) defined as the time from enrollment to discharge or a decline of two steps on the clinical seven-step ordinal scale, whichsoever came first. A total of 60 severely ill patients with positive RT-PCR and Chest CT scans underwent randomization (20 patients to each arm). In the Intention-To-Treat population, IFNβ1a was associated with a significant difference against the control group, in the TTCI; (HR; 2.36, 95% CI 1.10–5.17, P-value = 0.031) while the IFNβ1b indicated no significant difference compared with the control; HR; 1.42, (95% CI 0.63–3.16, P-value = 0.395). The median TTCI for both of the intervention groups was five days vs. seven days for the control group. The mortality was numerically lower in both of the intervention groups (20% in the IFNβ1a group and 30% in the IFNβ1b group vs. 45% in the control group). There were no significant differences between the three arms regarding the adverse events. In patients with laboratory-confirmed SARS-CoV-2 infection, as compared with the base therapeutic regiment, the benefit of a significant reduction in TTCI was observed in the IFNβ1a arm. This finding needs further confirmation in larger studies.
Trial Registration Number: ClinicalTrials.gov, NCT04343768. (Submitted: 08/04/2020; First Online: 13/04/2020) (Registration Number: NCT04343768).
Journal Article
The Hippo signaling pathway in leukemia: function, interaction, and carcinogenesis
by
Hayatmoghadam, Bentolhoda
,
Kavianpour, Maria
,
Golmohammadi, Maryam
in
Apoptosis
,
Biomedical and Life Sciences
,
Biomedicine
2021
Cancer can be considered as a communication disease between and within cells; nevertheless, there is no effective therapy for the condition, and this disease is typically identified at its late stage. Chemotherapy, radiation, and molecular-targeted treatment are typically ineffective against cancer cells. A better grasp of the processes of carcinogenesis, aggressiveness, metastasis, treatment resistance, detection of the illness at an earlier stage, and obtaining a better therapeutic response will be made possible. Researchers have discovered that cancerous mutations mainly affect signaling pathways. The Hippo pathway, as one of the main signaling pathways of a cell, has a unique ability to cause cancer. In order to treat cancer, a complete understanding of the Hippo signaling system will be required. On the other hand, interaction with other pathways like Wnt, TGF-β, AMPK, Notch, JNK, mTOR, and Ras/MAP kinase pathways can contribute to carcinogenesis. Phosphorylation of oncogene YAP and TAZ could lead to leukemogenesis, which this process could be regulated via other signaling pathways. This review article aimed to shed light on how the Hippo pathway interacts with other cellular signaling networks and its functions in leukemia.
Journal Article
Combining cell therapy with human autologous Schwann cell and bone marrow-derived mesenchymal stem cell in patients with subacute complete spinal cord injury: safety considerations and possible outcomes
by
Oraee-Yazdani, Maryam
,
Zali, Alireza
,
Golmohammadi, Maryam
in
Adverse events
,
Antibiotics
,
Autografts
2021
Background
Cellular transplantations have promising effects on treating spinal cord injury (SCI) patients. Mesenchymal stem cells (MSCs) and Schwann cells (SCs), which have safety alongside their complementary characteristics, are suggested to be the two of the best candidates in SCI treatment. In this study, we assessed the safety and possible outcomes of intrathecal co-transplantation of autologous bone marrow MSC and SC in patients with subacute traumatic complete SCI.
Methods
Eleven patients with complete SCI (American Spinal Injury Association Impairment Scale (AIS); grade A) were enrolled in this study during the subacute period of injury. The patients received an intrathecal autologous combination of MSC and SC and were followed up for 12 months. We assessed the neurological changes by the American Spinal Injury Association’s (ASIA) sensory-motor scale, functional recovery by spinal cord independence measure (SCIM-III), and subjective changes along with adverse events (AE) with our checklist. Furthermore, electromyography (EMG), nerve conduction velocity (NCV), magnetic resonance imaging (MRI), and urodynamic study (UDS) were conducted for all the patients at the baseline, 6 months, and 1 year after the intervention.
Results
Light touch AIS score alterations were approximately the same as the pinprick changes (11.6 ± 13.1 and 12 ± 13, respectively) in 50% of the cervical and 63% of the lumbar-thoracic patients, and both were more than the motor score alterations (9.5 ± 3.3 in 75% of the cervical and 14% of the lumbar-thoracic patients). SCIM III total scores (21.2 ± 13.3) and all its sub-scores (“respiration and sphincter management” (15 ± 9.9), “mobility” (9.5 ± 13.3), and “self-care” (6 ± 1.4)) had statistically significant changes after cell injection. Our findings support that the most remarkable positive, subjective improvements were in trunk movement, equilibrium in standing/sitting position, the sensation of the bladder and rectal filling, and the ability of voluntary voiding. Our safety evaluation revealed no systemic complications, and radiological images showed no neoplastic overgrowth, syringomyelia, or pseudo-meningocele.
Conclusion
The present study showed that autologous SC and bone marrow-derived MSC transplantation at the subacute stage of SCI could reveal statistically significant improvement in sensory and neurological functions among the patients. It appears that using this combination of cells is safe and effective for clinical application to spinal cord regeneration during the subacute period.
Journal Article
Intracerebral Administration of Autologous Mesenchymal Stem Cells as HSV-TK Gene Vehicle for Treatment of Glioblastoma Multiform: Safety and Feasibility Assessment
by
Rostami, Fatemeh
,
Jamshidi-Adegani, Fatemeh
,
Oraee-Yazdani, Maryam
in
Adult
,
Antineoplastic Agents - administration & dosage
,
Antineoplastic Agents - therapeutic use
2021
Widespread investigation has revealed the promising ability of suicidal genes in the treatment of glioma tumors; nevertheless, promoting their effects relies on the ability to apply suitable vehicles and techniques. In this study, the safety and feasibility of using bone marrow–derived mesenchymal stem cells (MSCs) in combination with prodrug for treatment of patients with primary and secondary glioblastoma multiform (GBM) was assessed. Five GBM patients were recruited. Following gross total resection of the tumor and adjuvant radiotherapy and chemotherapy, intracerebral injection of autologous MSCs transduced with lentivirus containing herpes simplex virus thymidine kinase (HSV-TK) was performed followed by intravenous administration of ganciclovir for 2 weeks. The treatment was well tolerated by all patients. Mild-to-moderate fever, headache, and cerebrospinal fluid leukocytosis were evident in three, two, and one patient, respectively. The progression-free survival (PFS) and overall survival (OS) of patients were 95.79 ± 51.40 and 128.85 ± 48.81 weeks, respectively. The 1-year PFS and OS were 60% and 100%, respectively, among our patients, and two patients had more than 3 years of OS and more than 2 years of PFS. It seems that intracerebral administration of bone marrow MSC containing the HSV-TK gene in combination with intravenous ganciclovir would be safe and feasible in the treatment of patients with GBM.
Journal Article
Effectiveness of Interferon Beta 1a, compared to Interferon Beta 1b and the usual therapeutic regimen to treat adults with moderate to severe COVID-19: structured summary of a study protocol for a randomized controlled trial
by
Golmohammadi, Maryam
,
Darazam, Ilad Alavi
,
Shokouhi, Shervin
in
Antiviral agents
,
Antiviral Agents - adverse effects
,
Antiviral Agents - therapeutic use
2020
Objectives
We will investigate the effectiveness of Interferon Beta 1a, compared to Interferon Beta 1b and the usual therapeutic regimen in COVID-19 in patients that have tested positive and are moderately to severely ill.
Trial design
This is a single center, open label, randomized, controlled, parallel group, clinical trial that will be conducted at Loghman Hakim Medical Education Center in conjunction with Shahid Beheshti University of Medical Sciences.
Participants
Sixty COVID-19 confirmed cases (using the RT-PCR test) will be enrolled in the trial between April 9
th
to April 14
th
2020. Patients will be randomly assigned to the intervention groups or the control group with the following eligibility criteria: ≥ 18 years of age AND (oxygen saturation (SPO2) ≤ 93% OR respiratory rate ≥ 24) AND at least one of the following: Contactless infrared forehead thermometer temperature of ≥37.8, cough, sputum production, nasal discharge, myalgia, headache or fatigue on admission, and time of onset of the symptoms should be acute (Days ≤ 14). Although Hydroxychloroquine will be administered in a single dose, patients with heart problems (prolonged QT or PR intervals, second- or third-degree heart block, and arrhythmias including torsade de pointes) will be excluded. Other exclusion criteria include using drugs with potential interaction with Hydroxychloroquine + Lopinavir/Ritonavir, Interferon-β 1a, Interferon-β 1b, pregnant or lactating women, history of alcohol or drug addiction in the past 5 years, blood ALT/AST levels > 5 times the upper limit of normal on laboratory results and refusal to participate.
This study will be undertaken at the Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences and Health Services.
Intervention and comparator
COVID-19 confirmed patients will be randomly assigned to one of three groups, with 20 patients in each. The first group (Arm 1) will receive Hydroxychloroquine + Lopinavir / Ritonavir (Kaletra) + Interferon-β 1a (Recigen), the second group (Arm 2) will be administered Hydroxychloroquine + Lopinavir / Ritonavir (Kaletra) + Interferon-β 1b (Ziferon), and the control group (Arm 3) will be treated by Hydroxychloroquine + Lopinavir / Ritonavir (Kaletra).
Main outcomes
Time to clinical improvement is our primary outcome measure. This is an improvement of two points on a seven-category ordinal scale (recommended by the World Health Organization: Coronavirus disease (COVID-2019) R&D. Geneva: World Health Organization) or discharge from the hospital, whichever comes first.
Secondary outcomes include mortality from the date of randomization until the last day of the study which will be the day all of the patients have had at least one of the following outcomes: 1) Improvement of two points on a seven-category ordinal scale. 2) Discharge from the hospital 3) Death. If any patient dies, we have reached an important secondary outcome. SpO2 Improvement between the last and first day of hospitalization, using pulse-oximetry. Duration of hospitalization from date of randomization until the date of hospital discharge or date of death from any cause, whichever comes first. Incidence of new mechanical ventilation uses from date of randomization until the last day of the study. Please note that we are trying to add further secondary outcomes and this section of the protocol is still evolving.
Statistical analysis will be performed by R version 3.6.1 software. We will use Kaplan–Meier to analyze the time to clinical improvement (compared with a log-rank test). Hazard ratios with 95% confidence intervals will be calculated using the Cox proportional-hazards model in crude and adjusted analysis.
Randomization
Eligible patients will be randomly assigned in a 1:1:1 ratio to receive either Interferon Beta 1a, Interferon Beta 1b or standard care only. Patients will be randomly allocated to three therapeutic arms using permuted, block-randomization to balance the number of patients allocated to each group. The permuted block (three or six patients per block) randomization sequence will be generated, using Package ‘randomizeR’ in R software version 3.6.1. and placed in individual sealed and opaque envelopes by the statistician. The investigator will enroll the patients and only then open envelopes to assign patients to the different treatment groups. This method of allocation concealment will result in minimum selection and confounding biases.
Blinding (Masking)
The present research is open-label (no masking) of patients and health care professionals who are undertaking outcome assessment of the primary outcome - time to clinical improvement.
Numbers to be Randomized (Sample Size)
Of the 60 patients who underwent randomization, 20 patients were assigned to receive Interferon beta-1a, 20 patients were assigned to receive Interferon beta 1b plus standard care and the rest of patients were assigned to receive the standard care alone.
Trial Status
Protocol version 1.2.1. Recruitment is finished, the start date of recruitment was on 9
th
April 2020 and the end date was on 14
th
April 2020. Last point of data collection will be the last day on which all of the 60 participants have had an outcome of clinical improvement or death, completing the study’s follow-up time window.
Trial registration
This study was registered with National Institutes of Health Clinical trials (
www.clinicaltrials.gov
; identification number NCT04343768, registered April 8, 2020 and first available online April 13, 2020).
Full protocol
The full protocol is attached as an additional file, accessible from the Trials website (Additional file
1
). In the interest in expediting dissemination of this material, the familiar formatting has been eliminated; this Letter serves as a summary of the key elements of the full protocol.
Journal Article
Protective effects of pioglitazone in renal ischemia–reperfusion injury (RIRI): focus on oxidative stress and inflammation
by
Alazbjee, Adeeb Abdulally Abdulhussien
,
Golmohammadi, Maryam
,
Mustafa, Yasser Fakri
in
Acute Kidney Injury - drug therapy
,
Acute Kidney Injury - metabolism
,
Acute Kidney Injury - prevention & control
2024
Background
Renal ischemia-reperfusion injury (RIRI) is a critical phenomenon that compromises renal function and is the most serious health concern related to acute kidney injury (AKI). Pioglitazone (Pio) is a known agonist of peroxisome proliferator-activated receptor-gamma (PPAR-γ). PPAR-γ is a nuclear receptor that regulates genes involved in inflammation, metabolism, and cellular differentiation. Activation of PPAR-γ is associated with antiinflammatory and antioxidant effects, which are relevant to the pathophysiology of RIRI. This study aimed to investigate the protective effects of Pio in RIRI, focusing on oxidative stress and inflammation.
Methods
We conducted a comprehensive literature search using electronic databases, including PubMed, ScienceDirect, Web of Science, Scopus, and Google Scholar.
Results
The results of this study demonstrated that Pio has antioxidant, anti-inflammatory, and anti-apoptotic activities that counteract the consequences of RIRI. The study also discussed the underlying mechanisms, including the modulation of various pathways such as TNF-α, NF-κB signaling systems, STAT3 pathway, KIM-1 and NGAL pathways, AMPK phosphorylation, and autophagy flux. Additionally, the study presented a summary of various animal studies that support the potential protective effects of Pio in RIRI.
Conclusion
Our findings suggest that Pio could protect the kidneys from RIRI by improving antioxidant capacity and decreasing inflammation. Therefore, these findings support the potential of Pio as a therapeutic strategy for preventing RIRI in different clinical conditions.
Graphical abstract
Pioglitazone (Pio) plays a protective role in renal ischemia–reperfusion injury (RIRI) by regulating oxidative stress and inflammation responses. In an animal model of RIRI, Pio reduced inflammation, and oxidative stress renal tubule damage through several cellular pathways. It also promotes cell viability and inhibits autophagy. Pioe targets various pathways such as TNF-α, NF-κB signaling systems, KIM-1 and NGAL pathways, and AMPK phosphorylation.
Journal Article
Targeting STAT3 signaling pathway by curcumin and its analogues for breast cancer: A narrative review
by
Aghaei, Zeinab Hashem
,
Golmohammadi, Maryam
,
Jabbar, Thaer L.
in
adjuvants
,
Animals
,
Antineoplastic Agents - pharmacology
2024
Background
Breast cancer (BC) continues to be a significant global health issue, with a rising number of cases requiring ongoing research and innovation in treatment strategies. Curcumin (CUR), a natural compound derived from Curcuma longa, and similar compounds have shown potential in targeting the STAT3 signaling pathway, which plays a crucial role in BC progression.
Aims
The aim of this study was to investigate the effects of curcumin and its analogues on BC based on cellular and molecular mechanisms.
Materials & Methods
The literature search conducted for this study involved utilizing the Scopus, ScienceDirect, PubMed, and Google Scholar databases in order to identify pertinent articles.
Results
This narrative review explores the potential of CUR and similar compounds in inhibiting STAT3 activation, thereby suppressing the proliferation of cancer cells, inducing apoptosis, and inhibiting metastasis. The review demonstrates that CUR directly inhibits the phosphorylation of STAT3, preventing its movement into the nucleus and its ability to bind to DNA, thereby hindering the survival and proliferation of cancer cells. CUR also enhances the effectiveness of other therapeutic agents and modulates the tumor microenvironment by affecting tumor‐associated macrophages (TAMs). CUR analogues, such as hydrazinocurcumin (HC), FLLL11, FLLL12, and GO‐Y030, show improved bioavailability and potency in inhibiting STAT3, resulting in reduced cell proliferation and increased apoptosis.
Conclusion
CUR and its analogues hold promise as effective adjuvant treatments for BC by targeting the STAT3 signaling pathway. These compounds provide new insights into the mechanisms of action of CUR and its potential to enhance the effectiveness of BC therapies.
The figure is a graphical illustrating the effects of curcumin and its derivatives on various breast cancer cell lines, including MCF‐7, MCF10A, and MDA‐MB‐231 cells. Curcumin is shown to inhibit the proliferation of MDA‐MB‐231 cells and enhance the effects of IFN‐β/RA on breast cancer cells by up‐regulating GRIM‐19. It also inhibits STAT3 activation in MCF10A cells and increases the ratio of Bax to Bcl‐2 proteins while decreasing the activation of STAT3 in MCF‐7 cells. Hydrazino curcumin specifically inhibits STAT3 activation in MCF‐7 cells. Additionally, FLLL11 and FLLL12 inhibit AKT and STAT3 phosphorylation in breast cancer cells.
Journal Article
Neurological, functional, and quality of life outcomes following combined mesenchymal stem cell and Schwann cell therapy in spinal cord injury: a 9-year experience
2025
Background
Spinal cord injury (SCI) often results in severe disabilities and significant socioeconomic burdens.
Objective
This study aimed to evaluate the effects and safety of co-transplantation of autologous bone marrow-derived mesenchymal stem cells (MSCs) and Schwann cells (SCs) via the intrathecal route in patients with complete spinal cord injury (SCI). The analysis focused on the therapy’s impact across various SCI subgroups (cervical vs. thoracolumbar, subacute vs. chronic) and the factors influencing its efficacy.
Methods
This case series evaluated 106 patients with complete SCI treated with combined cell therapy between August 2013 and September 2022, with a one-year follow-up. Safety profiles were assessed, and neurological and functional outcomes were measured using the American Spinal Injury Association (ASIA) scores, Spinal Cord Independence Measure (SCIM-III), and the World Health Organization Quality of Life Brief Version (WHOQOL-BREF) at 6- and 12-month intervals post-injection. Multiple regression analysis was conducted to evaluate factors associated with outcomes.
Results
Significant improvements were observed in ASIA scores (motor, light touch, and pinprick), SCIM-III scores (total and subscales), and WHOQOL-BREF scores after 12 months. These improvements were consistent across subgroups, regardless of injury level or duration. Multiple regression analysis indicated that improvements in ASIA motor scores were associated with injury level, while improvements in SCIM-III total and mobility scores were associated with time since injury and patient age.
Conclusions
This study demonstrates significant neurological, functional, and quality of life improvements following combined cell therapy with autologous MSCs and SCs in patients with complete SCI. Future research should investigate potential synergies with other therapies and conduct comparative efficacy analyses.
Graphical abstract
Journal Article
Metformin may alter the course of Leber’s hereditary optic neuropathy: a case report
by
Sadun, Alfredo A.
,
Gauthier, Danielle A.
,
Golmohammadi, Maryam
in
Diabetes
,
Disease
,
Hormone replacement therapy
2025
Leber’s hereditary optic neuropathy (LHON) is a rare inherited mitochondrial disease caused by variants in mitochondrial DNA (mtDNA) transmitted exclusively through the maternal line. The disease predominantly affects young males and is characterized by progressive bilateral vision loss. Idebenone, a well-studied drug, modestly enhances the mitochondrial function and visual acuity in many patients with LHON. In this study, we report the case of a 48-year-old woman diagnosed with LHON (m.11778G>A/ MT-ND4 ) and type 2 diabetes mellitus who experienced visual field improvement following metformin treatment after 26 months of progressive vision loss unresponsive to idebenone, nicotinamide adenine dinucleotide (NAD+), and hormone replacement therapy (HRT). Our findings offer an intriguing perspective on LHON management but require more investigations, particularly on the molecular effects of metformin on the mitochondrial function in LHON patients.
Journal Article
Improving dexamethasone drug loading and efficacy in treating rheumatoid arthritis via liposome: Focusing on inflammation and molecular mechanisms
by
Nikbakht, Nikta
,
Golmohammadi, Maryam
,
Skakodub, Alla A.
in
Animal models
,
Animals
,
Anti-Inflammatory Agents - administration & dosage
2025
Rheumatoid arthritis (RA) is a chronic autoimmune disease that affects approximately 0.46% of the global population. Conventional therapeutics for RA, including disease‐modifying antirheumatic drugs (DMARDs), nonsteroidal anti‐inflammatory drugs (NSAIDs), and corticosteroids, frequently result in unintended adverse effects. Dexamethasone (DEX) is a potent glucocorticoid used to treat RA due to its anti‐inflammatory and immunosuppressive properties. Liposomal delivery of DEX, particularly when liposomes are surface‐modified with targeting ligands like peptides or sialic acid, can improve drug efficacy by enhancing its distribution to inflamed joints and minimizing toxicity. This study investigates the potential of liposomal drug delivery systems to enhance the efficacy and targeting of DEX in the treatment of RA. Results from various studies demonstrate that liposomal DEX significantly inhibits arthritis progression in animal models, reduces joint inflammation and damage, and alleviates cartilage destruction compared to free DEX. The liposomal formulation also shows better hemocompatibility, fewer adverse effects on body weight and immune organ index, and a longer circulation time with higher bioavailability. The anti‐inflammatory mechanism is associated with the downregulation of pro‐inflammatory cytokines like tumor necrosis factor‐α (TNF‐α) and B‐cell–activating factor (BAFF), which are key players in the pathogenesis of RA. Additionally, liposomal DEX can induce the expression of anti‐inflammatory cytokines like interleukin‐10 (IL‐10), which has significant anti‐inflammatory and immunoregulatory properties. The findings suggest that liposomal DEX represents a promising candidate for effective and safe RA therapy, with the potential to improve the management of this debilitating disease by providing targeted delivery and sustained release of the drug.
Liposomal dexamethasone formulations offer targeted delivery and enhanced efficacy in treating rheumatoid arthritis (RA). Dex@FA‐ROS‐Lips utilize folic acid targeting to accumulate in inflamed joints, reducing cartilage destruction and joint swelling while improving circulation time and bioavailability. hPG‐Dex liposomes enhance regulatory T cells and increase IL‐10 expression, leading to systemic tolerance and reduced arthritis progression. Polymerized stealth liposomes preferentially accumulate in inflamed joints, decreasing TNF‐α and IL‐1β levels, thus mitigating arthritis symptoms. DP‐SAL liposomes target neutrophils via sialic acid modification, enhancing anti‐inflammatory effects and drug accumulation in joints. Dex‐Lips reduce paw swelling, TNF‐α, and IL‐1β levels, showing superior therapeutic efficacy and fewer side effects compared to free DEX. These liposomal formulations demonstrate significant potential in improving RA treatment by providing targeted delivery and sustained release of DEX. Future research should focus on optimizing particle size, exploring advanced targeting strategies, and addressing scalability and regulatory challenges. Additionally, combining liposomal DEX with other therapies could further enhance treatment outcomes. Overall, liposomal DEX represents a promising approach for effective and safe RA therapy.
Journal Article