Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ajaikumar B. Kunnumakkara is active.

Publication


Featured researches published by Ajaikumar B. Kunnumakkara.


Cancer Letters | 2008

Curcumin inhibits proliferation, invasion, angiogenesis and metastasis of different cancers through interaction with multiple cell signaling proteins

Ajaikumar B. Kunnumakkara; Preetha Anand; Bharat B. Aggarwal

Because most cancers are caused by dysregulation of as many as 500 different genes, agents that target multiple gene products are needed for prevention and treatment of cancer. Curcumin, a yellow coloring agent in turmeric, has been shown to interact with a wide variety of proteins and modify their expression and activity. These include inflammatory cytokines and enzymes, transcription factors, and gene products linked with cell survival, proliferation, invasion, and angiogenesis. Curcumin has been found to inhibit the proliferation of various tumor cells in culture, prevents carcinogen-induced cancers in rodents, and inhibits the growth of human tumors in xenotransplant or orthotransplant animal models either alone or in combination with chemotherapeutic agents or radiation. Several phase I and phase II clinical trials indicate that curcumin is quite safe and may exhibit therapeutic efficacy. These aspects of curcumin are discussed further in detail in this review.


Cancer Letters | 2008

Curcumin and cancer: An “old-age” disease with an “age-old” solution

Preetha Anand; Chitra Sundaram; Sonia Jhurani; Ajaikumar B. Kunnumakkara; Bharat B. Aggarwal

Cancer is primarily a disease of old age, and that life style plays a major role in the development of most cancers is now well recognized. While plant-based formulations have been used to treat cancer for centuries, current treatments usually involve poisonous mustard gas, chemotherapy, radiation, and targeted therapies. While traditional plant-derived medicines are safe, what are the active principles in them and how do they mediate their effects against cancer is perhaps best illustrated by curcumin, a derivative of turmeric used for centuries to treat a wide variety of inflammatory conditions. Curcumin is a diferuloylmethane derived from the Indian spice, turmeric (popularly called curry powder) that has been shown to interfere with multiple cell signaling pathways, including cell cycle (cyclin D1 and cyclin E), apoptosis (activation of caspases and down-regulation of antiapoptotic gene products), proliferation (HER-2, EGFR, and AP-1), survival (PI3K/AKT pathway), invasion (MMP-9 and adhesion molecules), angiogenesis (VEGF), metastasis (CXCR-4) and inflammation (NF-kappaB, TNF, IL-6, IL-1, COX-2, and 5-LOX). The activity of curcumin reported against leukemia and lymphoma, gastrointestinal cancers, genitourinary cancers, breast cancer, ovarian cancer, head and neck squamous cell carcinoma, lung cancer, melanoma, neurological cancers, and sarcoma reflects its ability to affect multiple targets. Thus an old-age disease such as cancer requires an age-old treatment.


Annals of the New York Academy of Sciences | 2009

Signal Transducer and Activator of Transcription‐3, Inflammation, and Cancer

Bharat B. Aggarwal; Ajaikumar B. Kunnumakkara; Kuzhuvelil B. Harikumar; Shan R. Gupta; Sheeja T. Tharakan; Cemile Koca; Sanjit Dey; Bokyung Sung

Signal transducer and activator of transcription‐3 (STAT‐3) is one of six members of a family of transcription factors. It was discovered almost 15 years ago as an acute‐phase response factor. This factor has now been associated with inflammation, cellular transformation, survival, proliferation, invasion, angiogenesis, and metastasis of cancer. Various types of carcinogens, radiation, viruses, growth factors, oncogenes, and inflammatory cytokines have been found to activate STAT‐3. STAT‐3 is constitutively active in most tumor cells but not in normal cells. Phosphorylation of STAT‐3 at tyrosine 705 leads to its dimerization, nuclear translocation, DNA binding, and gene transcription. The phosphorylation of STAT‐3 at serine 727 may regulate its activity negatively or positively. STAT‐3 regulates the expression of genes that mediate survival (survivin, bcl‐xl, mcl‐1, cellular FLICE‐like inhibitory protein), proliferation (c‐fos, c‐myc, cyclin D1), invasion (matrix metalloproteinase‐2), and angiogenesis (vascular endothelial growth factor). STAT‐3 activation has also been associated with both chemoresistance and radioresistance. STAT‐3 mediates these effects through its collaboration with various other transcription factors, including nuclear factor‐κB, hypoxia‐inducible factor‐1, and peroxisome proliferator activated receptor‐γ. Because of its critical role in tumorigenesis, inhibitors of this factors activation are being sought for both prevention and therapy of cancer. This has led to identification of small peptides, oligonucleotides, and small molecules as potential STAT‐3 inhibitors. Several of these small molecules are chemopreventive agents derived from plants. This review discusses the intimate relationship between STAT‐3, inflammation, and cancer in more detail.


Cancer Research | 2007

Curcumin Potentiates Antitumor Activity of Gemcitabine in an Orthotopic Model of Pancreatic Cancer through Suppression of Proliferation, Angiogenesis, and Inhibition of Nuclear Factor-κB–Regulated Gene Products

Ajaikumar B. Kunnumakkara; Sushovan Guha; Sunil Krishnan; Parmeswaran Diagaradjane; Juri G. Gelovani; Bharat B. Aggarwal

Gemcitabine is currently the best treatment available for pancreatic cancer, but the disease develops resistance to the drug over time. Agents that can either enhance the effects of gemcitabine or overcome chemoresistance to the drug are needed for the treatment of pancreatic cancer. Curcumin, a component of turmeric (Curcuma longa), is one such agent that has been shown to suppress the transcription factor nuclear factor-kappaB (NF-kappaB), which is implicated in proliferation, survival, angiogenesis, and chemoresistance. In this study, we investigated whether curcumin can sensitize pancreatic cancer to gemcitabine in vitro and in vivo. In vitro, curcumin inhibited the proliferation of various pancreatic cancer cell lines, potentiated the apoptosis induced by gemcitabine, and inhibited constitutive NF-kappaB activation in the cells. In vivo, tumors from nude mice injected with pancreatic cancer cells and treated with a combination of curcumin and gemcitabine showed significant reductions in volume (P = 0.008 versus control; P = 0.036 versus gemcitabine alone), Ki-67 proliferation index (P = 0.030 versus control), NF-kappaB activation, and expression of NF-kappaB-regulated gene products (cyclin D1, c-myc, Bcl-2, Bcl-xL, cellular inhibitor of apoptosis protein-1, cyclooxygenase-2, matrix metalloproteinase, and vascular endothelial growth factor) compared with tumors from control mice treated with olive oil only. The combination treatment was also highly effective in suppressing angiogenesis as indicated by a decrease in CD31(+) microvessel density (P = 0.018 versus control). Overall, our results suggest that curcumin potentiates the antitumor effects of gemcitabine in pancreatic cancer by suppressing proliferation, angiogenesis, NF-kappaB, and NF-kappaB-regulated gene products.


Biochemical Pharmacology | 2010

Design of curcumin-loaded PLGA nanoparticles formulation with enhanced cellular uptake, and increased bioactivity in vitro and superior bioavailability in vivo.

Preetha Anand; Hareesh B. Nair; Bokyung Sung; Ajaikumar B. Kunnumakkara; Vivek R. Yadav; Rajeshwar Rao Tekmal; Bharat B. Aggarwal

Curcumin, a yellow pigment present in the spice turmeric (Curcuma longa), has been linked with antioxidant, anti-inflammatory, antiproliferative, anticancer, antidiabetic, antirheumatic, and antiviral effects, but its optimum potential is limited by its lack of solubility in aqueous solvents and poor oral bioavailability. We employed a polymer-based nanoparticle approach to improve bioavailability. Curcumin was encapsulated with 97.5% efficiency in biodegradable nanoparticulate formulation based on poly (lactide-co-glycolide) (PLGA) and a stabilizer polyethylene glycol (PEG)-5000. Dynamic laser light scattering and transmission electron microscopy indicated a particle diameter of 80.9 nm. This curcumin, renamed from hereon as curcumin (NP), was characterized for its biological activity. In vitro curcumin (NP) exhibited very rapid and more efficient cellular uptake than curcumin. Estrase staining revealed that curcumin (NP) was at least as potent as or more potent than curcumin in inducing apoptosis of leukemic cells and in suppressing proliferation of various tumor cell lines. When examined by electrophoretic gel shift mobility assay, curcumin (NP) was more active than curcumin in inhibiting TNF-induced NF-kappaB activation and in suppression of NF-kappaB-regulated proteins involved in cell proliferation (cyclin D1), invasion (MMP-9), and angiogenesis (VEGF). In mice, curcumin (NP) was more bioavailable and had a longer half-life than curcumin. Overall we demonstrate that curcumin-loaded PLGA nanoparticles formulation has enhanced cellular uptake, and increased bioactivity in vitro and superior bioavailability in vivo over curcumin.


Annals of the New York Academy of Sciences | 2006

Targeting Signal-Transducer-and-Activator-of-Transcription-3 for Prevention and Therapy of Cancer

Bharat B. Aggarwal; Gautam Sethi; Kwang Seok Ahn; Santosh K. Sandur; Manoj Pandey; Ajaikumar B. Kunnumakkara; Bokyung Sung; Haruyo Ichikawa

Abstract:u2002 Recent evidence indicates a convergence of molecular targets for both prevention and therapy of cancer. Signal‐transducer‐and‐activator‐of‐transcription‐3 (STAT3), a member of a family of six different transcription factors, is closely linked with tumorigenesis. Its role in cancer is indicated by numerous avenues of evidence, including the following: STAT3 is constitutively active in tumor cells; STAT3 is activated by growth factors (e.g., EGF, TGF‐α, IL‐6, hepatocyte growth factor) and oncogenic kinases (e.g., Src); STAT3 regulates the expression of genes that mediate proliferation (e.g., c‐myc and cyclin D1), suppress apoptosis (e.g., Bcl‐xL and survivin), or promote angiogenesis (e.g, VEGF); STAT3 activation has been linked with chemoresistance and radioresistance; and chemopreventive agents have been shown to suppress STAT3 activation. Thus inhibitors of STAT3 activation have potential for both prevention and therapy of cancer. Besides small peptides and oligonucleotides, numerous small molecules have been identified as blockers of STAT3 activation, including synthetic molecules (e.g., AG 490, decoy peptides, and oligonucleotides) and plant polyphenols (e.g., curcumin, resveratrol, flavopiridol, indirubin, magnolol, piceatannol, parthenolide, EGCG, and cucurbitacin). This article discusses these aspects of STAT3 in more detail.


Clinical Cancer Research | 2007

Curcumin Inhibits Tumor Growth and Angiogenesis in Ovarian Carcinoma by Targeting the Nuclear Factor-κB Pathway

Yvonne G. Lin; Ajaikumar B. Kunnumakkara; Asha S. Nair; William M. Merritt; Liz Y. Han; Guillermo N. Armaiz-Pena; Aparna A. Kamat; Whitney A. Spannuth; David M. Gershenson; Susan K. Lutgendorf; Bharat B. Aggarwal; Anil K. Sood

Purpose: Curcumin, a component of turmeric, has been shown to suppress inflammation and angiogenesis largely by inhibiting the transcription factor nuclear factor-κB (NF-κB). This study evaluates the effects of curcumin on ovarian cancer growth using an orthotopic murine model of ovarian cancer. Experimental Design:In vitro and in vivo experiments of curcumin with and without docetaxel were done using human ovarian cancer cell lines SKOV3ip1, HeyA8, and HeyA8-MDR in athymic mice. NF-κB modulation was ascertained using electrophoretic mobility shift assay. Evaluation of angiogenic cytokines, cellular proliferation (proliferating cell nuclear antigen), angiogenesis (CD31), and apoptosis (terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling) was done using immunohistochemical analyses. Results: Curcumin inhibited inducible NF-κB activation and suppressed proliferation in vitro. In vivo dose-finding experiments revealed that 500 mg/kg orally was the optimal dose needed to suppress NF-κB and signal transducers and activators of transcription 3 activation and decrease angiogenic cytokine expression. In the SKOV3ip1 and HeyA8 in vivo models, curcumin alone resulted in 49% (P = 0.08) and 55% (P = 0.01) reductions in mean tumor growth compared with controls, whereas when combined with docetaxel elicited 96% (P < 0.001) and 77% reductions in mean tumor growth compared with controls. In mice with multidrug-resistant HeyA8-MDR tumors, treatment with curcumin alone and combined with docetaxel resulted in significant 47% and 58% reductions in tumor growth, respectively (P = 0.05). In SKOV3ip1 and HeyA8 tumors, curcumin alone and with docetaxel decreased both proliferation (P < 0.001) and microvessel density (P < 0.001) and increased tumor cell apoptosis (P < 0.05). Conclusions: Based on significant efficacy in preclinical models, curcumin-based therapies may be attractive in patients with ovarian carcinoma.


Planta Medica | 2008

Potential of spice-derived phytochemicals for cancer prevention

Bharat B. Aggarwal; Ajaikumar B. Kunnumakkara; Kuzhuvelil B. Harikumar; Sheeja T. Tharakan; Bokyung Sung; Preetha Anand

Although spices have been used for thousands of years and are known for their flavor, taste and color in the food, they are not usually recognized for their medicinal value. Extensive research within the last two decades from our laboratory and others has indicated that there are phytochemicals present in spices that may prevent various chronic illnesses including cancerous, diabetic, cardiovascular, pulmonary, neurological and autoimmune diseases. For instance, the potential of turmeric (curcumin), red chilli (capsaicin), cloves (eugenol), ginger (zerumbone), fennel (anethole), kokum (gambogic acid), fenugreek (diosgenin), and black cumin (thymoquinone) in cancer prevention has been established. Additionally, the mechanism by which these agents mediate anticancer effects is also becoming increasingly evident. The current review describes the active components of some of the major spices, their mechanisms of action and their potential in cancer prevention.


Cancer Research | 2006

Overexpression of Tissue Transglutaminase Leads to Constitutive Activation of Nuclear Factor-κB in Cancer Cells: Delineation of a Novel Pathway

Aman P. Mann; Amit Verma; Gautam Sethi; Bramanandam Manavathi; Huamin Wang; Jansina Y. Fok; Ajaikumar B. Kunnumakkara; Rakesh Kumar; Bharat B. Aggarwal; Kapil Mehta

The transcription factor nuclear factor-kappaB (NF-kappaB) plays an important role in regulating cell growth, apoptosis, and metastatic functions. Constitutive activation of NF-kappaB has been observed in various cancers; however, molecular mechanisms resulting in such activation remain elusive. Based on our previous results showing that drug-resistant and metastatic cancer cells have high levels of tissue transglutaminase (TG2) expression and that this expression can confer chemoresistance to certain types of cancer cells, we hypothesized that TG2 contributes to constitutive activation of NF-kappaB. Numerous lines of evidence showed that overexpression of TG2 is linked with constitutive activation of NF-kappaB. Tumor cells with overexpression of TG2 exhibited increased levels of constitutively active NF-kappaB. Activation of TG2 led to activation of NF-kappaB; conversely, inhibition of TG2 activity inhibited activation of NF-kappaB. Similarly, ectopic expression of TG2 caused activation of NF-kappaB, and inhibition of expression of TG2 by small interfering RNA abolished the activation of NF-kappaB. Our results further indicated that constitutive NF-kappaB reporter activity in pancreatic cancer cells is not affected by dominant-negative I kappaB alpha. Additionally, coimmunoprecipitation and confocal microscopy showed that I kappaB alpha is physically associated with TG2. Lastly, immunohistochemical analysis of pancreatic ductal carcinoma samples obtained from 61 patients further supported a strong correlation between TG2 expression and NF-kappaB activation/overexpression (P = 0.0098, Fishers exact test). We conclude that TG2 induces constitutive activation of NF-kappaB in tumor cells via a novel pathway that is most likely independent of I kappaB alpha kinase. Therefore, TG2 may be an attractive alternate target for inhibiting constitutive NF-kappaB activation and rendering cancer cells sensitive to anticancer therapies.


Clinical Cancer Research | 2007

Capsaicin is a novel blocker of constitutive and interleukin-6 - Inducible STAT3 activation

Manisha Bhutani; Ashutosh K. Pathak; Asha S. Nair; Ajaikumar B. Kunnumakkara; Sushovan Guha; Gautam Sethi; Bharat B. Aggarwal

Purpose: Capsaicin, a constituent of green and red peppers, has been linked with suppression of tumorigenesis through a mechanism that is not well understood. Because the transcription factor signal transducer and activator of transcription 3 (STAT3) has been closely linked with tumorigenesis, we investigated the effect of this vanilloid on the STAT3 pathway in human multiple myeloma cells. Experimental Design: The effect of capsaicin on both constitutive and interleukin-6–induced STAT3 activation, associated protein kinases, and STAT3-regulated gene products involved in proliferation, survival and angiogenesis, cellular proliferation, and apoptosis in multiple myeloma cells was investigated. Results: We found that capsaicin inhibited constitutive activation of STAT3 in multiple myeloma cells in a dose- and time-dependent manner, with minimum effect on STAT5. Capsaicin also inhibited the interleukin-6–induced STAT3 activation. The activation of Janus-activated kinase 1 and c-Src, implicated in STAT3 activation, was also inhibited by the vanilloid, with no effect on extracellular signal-regulated kinase 1/2 activation. Pervanadate reversed the capsaicin-induced down-regulation of STAT3, suggesting the involvement of a protein tyrosine phosphatase. Capsaicin down-regulated the expression of the STAT3-regulated gene products, such as cyclin D1, Bcl-2, Bcl-xL, survivin, and vascular endothelial growth factor. Finally, capsaicin induced the accumulation of cells in G1 phase, inhibited proliferation, and induced apoptosis, as indicated by caspase activation. Capsaicin also significantly potentiated the apoptotic effects of Velcade and thalidomide in multiple myeloma cells. When administered i.p., capsaicin inhibited the growth of human multiple myeloma xenograft tumors in male athymic nu/nu mice. Conclusion: Overall, these results suggest that capsaicin is a novel blocker of the STAT3 activation pathway, with a potential role in the prevention and treatment of multiple myeloma and other cancers.

Collaboration


Dive into the Ajaikumar B. Kunnumakkara's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bokyung Sung

Pusan National University

View shared research outputs
Top Co-Authors

Avatar

Preetha Anand

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sushovan Guha

University of Texas System

View shared research outputs
Top Co-Authors

Avatar

Gautam Sethi

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Parmeswaran Diagaradjane

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sunil Krishnan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Juri G. Gelovani

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kuzhuvelil B. Harikumar

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge